Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 61(16): 6255-63, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507080

ABSTRACT

Interleukin-2-based regimens of biological therapy have shown some clinical promise for the treatment of kidney cancer in humans, although the mechanisms responsible for tumor regression occurring in these patients remain unclear. Preclinical insight into these mechanisms is limited by a paucity of orthotopic animal models of kidney cancer. We have used streptozotocin, an antibiotic and diabetogenic nitrosamine compound derived from Streptomyces achromogenes to induce new kidney tumors in BALB/c mice. Single or multiple doses of streptozotocin induced kidney tumors in up to 25% of mice by 50-90 weeks of age, with up to 18% characterized as renal cell carcinomas (RCCs). Several transplantable lines were obtained from the RCCs, and one of these lines was subsequently cloned. The initial tumor isolates and sublines were histologically reconfirmed to be RCCs, and all grew progressively but slowly (mean survival times, 57 to >100 days) in vivo after intrarenal implant. None of the primary isolates or sublines revealed mutations in either the VHL or Ras genes, although karyotype analysis and chromosome painting revealed the consistent presence of a submetacentric chromosome resulting from the fusion of chromosomes 16 and 19. Biological characterization of these tumors revealed several features analogous to the growth of human kidney cancers, including a propensity for the formation of lung metastases and high vascularity. This hypervascularity is evident by both gross and microscopic analysis and correlates with the expression of several proangiogenic genes. Overall, the features of orthotopic transplantability, slower in vivo growth (relative to the rapid growth rates of other transplantable mouse kidney tumors), propensity for lung metastases, and hypervascularity may make these tumors valuable models for the study of new therapeutic strategies based on antineovascular agents and antitumor cytokines.


Subject(s)
Carcinoma, Renal Cell/chemically induced , Kidney Neoplasms/chemically induced , Neoplasm Proteins , Streptozocin , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Division/drug effects , Cysteine Endopeptidases , DNA Mutational Analysis , Female , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Pathologic/genetics
2.
J Immunol ; 166(6): 3763-70, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238618

ABSTRACT

The IFN-gamma-inducible proteins monokine induced by IFN-gamma (Mig) and chemokine responsive to gamma-2 (Crg-2) can contribute to IL-12-induced antiangiogenic and leukocyte-recruiting activities, but the extent to which leukocytes vs parenchymal cells in different organs contribute to the production of these molecules remains unclear. The results presented herein show that IFN-gamma-dependent induction of Mig and Crg-2 gene expression can occur in many nonlymphoid organs, and these genes are rapidly induced in purified hepatocytes isolated from mice treated with IL-2 plus IL-12, or from Hepa 1-6 hepatoma cells treated in vitro with IFN-gamma. In addition to depending on IFN-gamma, the ability of IL-12 or IL-2/IL-12 to induce Mig and Crg-2 gene expression in purified hepatocytes also is accompanied by the coordinate up-regulation of the IFN-gamma R alpha and beta-chains, in the absence of IL-12R components. Supernatants of primary hepatocytes obtained from mice treated in vivo with IL-2/IL-12 or from hepatocytes treated in vitro with IFN-gamma contain increased chemotactic activity for enriched human and mouse CD3(+) T cells, as well as mouse DX5(+) NK cells. The hepatocyte-derived chemotactic activity for mouse T cells but not NK cells was ablated by Abs specific for Mig and Crg-2. These results suggest that parenchymal cells in some organs may contribute substantially to initiation and/or amplification of inflammatory or antitumor responses.


Subject(s)
Chemokines, CXC/physiology , Chemotaxis, Leukocyte/immunology , Hepatocytes/immunology , Hepatocytes/metabolism , Interleukin-12/administration & dosage , Interleukin-2/administration & dosage , Monokines/physiology , T-Lymphocytes/immunology , Animals , Cell Separation , Chemokine CXCL10 , Chemokine CXCL9 , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Chemotaxis, Leukocyte/genetics , Drug Combinations , Gene Expression Regulation/immunology , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Killer Cells, Natural/immunology , Liver/anatomy & histology , Liver/cytology , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Monokines/biosynthesis , Monokines/genetics , Receptors, Cytokine/biosynthesis , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Cells, Cultured
3.
J Immunol ; 166(2): 1156-68, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11145697

ABSTRACT

Using a novel transgenic mouse model of spontaneous mammary carcinoma, we show here that the IL-12/pulse IL-2 combination can induce rapid and complete regression of well-established autochthonous tumor in a setting where the host immune system has been conditioned by the full dynamic process of neoplastic progression and tumorigenesis. Further, this regimen inhibits neovascularization of established mammary tumors, and does so in conjunction with potent local induction of genes encoding the IFN-gamma- and TNF-alpha-inducible antiangiogenic chemokines IFN-inducible protein 10 and monokine induced by IFN-gamma. In contrast to untreated juvenile C3(1)TAg mice in which histologically normal mammary epithelium predictably undergoes progressive hyperplasia, atypical changes, and ultimately transition to overt carcinoma, the current studies also demonstrate a unique preventative therapeutic role for IL-12/pulse IL-2. In juvenile mice, early administration of IL-12/pulse IL-2 markedly limits the expected genetically programmed neoplastic transition within the mammary epithelium and does so in conjunction with enhancement of constitutive Fas and pronounced induction of local Fas ligand gene expression, T cell infiltration, and induction of apoptosis within the mammary epithelium. These events occur in the absence of a durable Ag-specific memory response. Thus, this novel model system demonstrates that the potent therapeutic activity of the IL-12/pulse IL-2 combination rapidly engages potent apoptotic and antiangiogenic mechanisms that remain active during the delivery of IL-12/pulse IL-2. The results also demonstrate that these mechanisms are active against established tumor as well as developing preneoplastic lesions.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/immunology , Cell Transformation, Neoplastic/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , fas Receptor/genetics , Age Factors , Angiogenesis Inhibitors/biosynthesis , Animals , Apoptosis/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Chemokines/biosynthesis , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Fas Ligand Protein , Female , Gene Expression Regulation, Neoplastic/immunology , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Interferon-gamma/physiology , Interleukin-12/administration & dosage , Interleukin-2/administration & dosage , Ligands , Lymphocytes, Tumor-Infiltrating/pathology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Remission Induction , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/immunology , fas Receptor/biosynthesis
4.
J Immunol ; 152(12): 5776-84, 1994 Jun 15.
Article in English | MEDLINE | ID: mdl-8207207

ABSTRACT

These studies investigate the effects of exogenously administered recombinant human IL-7 (rhIL-7) on mouse leukocyte subsets in vivo in normal and tumor-bearing mice. The administration of rhIL-7 to normal mice caused a pronounced leukocytosis (three- to fivefold increase over background) in the spleen and lymph nodes, with B-lineage and T cells, NK cells, and macrophages all being increased. CD8+ T cells increased disproportionately, such that the CD4 to CD8 ratio decreased dramatically. The rhIL-7-induced effects were dose-dependent, increased with duration of treatment, and were reversible after cessation of rhIL-7 administration. T cell number increases after rhIL-7 treatment were primarily a result of an expansion of the peripheral T cell population. Importantly, splenocytes from rhIL-7-treated mice have enhanced proliferative responses to various T cell stimuli in vitro and were able to potentiate an allogeneic CTL response in vivo. The rhIL-7-induced changes in T cell number and the CD4 to CD8 ratio also were observed in mice bearing early Renca renal adenocarcinoma pulmonary metastases, and these changes coincided with up to a 75% reduction in pulmonary metastases. Overall, these results demonstrate that the administration of rhIL-7 to mice profoundly increases the number of B and T cells, and reduces the number of pulmonary metastases. The results also suggest that IL-7 may be useful for restoring lymphoid subsets in immunosuppressed hosts and in enhancing T cell-mediated immune responses. Such effects may be useful in the treatment of microbial diseases and cancer.


Subject(s)
B-Lymphocytes/immunology , Interleukin-7/pharmacology , Neoplasm Metastasis/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, Differentiation/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/secondary , Carcinoma, Renal Cell/therapy , Cytotoxicity, Immunologic , Dose-Response Relationship, Immunologic , Humans , Interleukin-7/administration & dosage , Kidney Neoplasms/immunology , Kidney Neoplasms/secondary , Kidney Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Spleen/immunology
6.
Cancer Res ; 52(15): 4082-9, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1638519

ABSTRACT

Previous studies have demonstrated that interleukin 1 (IL-1) can protect most mice from the acute lethal toxicity mediated by high doses of radiation and/or some chemotherapeutic drugs. The results presented herein demonstrate that the pretreatment of mice with recombinant human interleukin 1 alpha (rhIL-1 alpha) protects mice from the lethal effects of several myelotoxic chemotherapeutic drugs, including 5-fluorouracil (5FUra), cyclophosphamide, cis-diammine(1,1-cyclobutanedicarboxylato)platinum(II), and 1,3-bis-(2-chloroethyl)-1-nitrosourea. However, pretreatment with rhIL-1 alpha was not effective against the acute lethal toxicity generated by doxorubicin and cisplatin. The chemoprotective effects appear to be at least partially due to myeloprotection/restoration, since the recovery of myeloid colony-forming units and the total cellularity in the bone marrow and spleen were accelerated in the rhIL-1 alpha-pretreated mice. However, the chemoprotective effects of rhIL-1 alpha are apparently not limited to myeloprotection, since pretreatment with rhIL-1 alpha protects mice against the lethal toxicity of both 5FUra and cyclophosphamide, yet bone marrow transplants rescue mice treated with 5FUra but not those treated with cyclophosphamide. The chemoprotective effects of rhIL-1 alpha may be at least partially indirect, since the efficacy of chemoprotection by rhIL-1 alpha is reduced in athymic mice, and interleukin 6, but not tumor necrosis factor alpha, can substitute for rhIL-1 alpha in chemoprotection from 5FUra.


Subject(s)
Antineoplastic Agents/toxicity , Bone Marrow/drug effects , Hematopoietic Stem Cells/drug effects , Interleukin-1/therapeutic use , Spleen/drug effects , Animals , Bone Marrow/pathology , Carboplatin/toxicity , Carmustine/toxicity , Cisplatin/toxicity , Cyclophosphamide/toxicity , Death , Doxorubicin/toxicity , Fluorouracil/toxicity , Hematopoietic Stem Cells/pathology , Humans , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Spleen/pathology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
7.
Cancer Res ; 51(24): 6596-602, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1742732

ABSTRACT

Flavone-8-acetic acid (FAA) is a flavonoid drug that augments mouse natural killer activity, induces cytokine gene expression, and synergizes with recombinant interleukin 2 for the treatment of murine renal cancer. However, FAA has been largely inactive in human clinical trials. In the present study we investigated the ability of FAA treatment to directly induce cytokine mRNA expression in total mouse splenic leukocytes and selected leukocyte subsets, as well as in total human peripheral blood leukocytes. Analysis of RNA isolated from FAA-treated mouse splenic leukocytes demonstrated that treatment with greater than or equal to 100 micrograms/ml of FAA induced expression of tumor necrosis factor alpha (TNF-alpha) mRNA by 1 h and induced maximal expression of TNF-alpha, alpha-interferon, and gamma-interferon mRNA within 3 h. The expression of all cytokine genes was diminished by 6 h. Interferon biological activity was detected in the supernatants of mouse splenic or peripheral blood leukocytes after treatment with FAA. These results correlate well with the previously reported induction of cytokine mRNA genes and biological activity by FAA in vivo. In contrast, FAA did not induce detectable mRNA expression or cytokine protein secretion by human peripheral blood leukocytes under similar conditions. These results demonstrate that FAA can directly stimulate cytokine gene expression in mouse but not in human leukocytes. Further studies performed with highly purified positively selected mouse CD4+ or CD8+ splenic T-lymphocytes, as well as purified B-cells, demonstrated that the FAA-induced expression of gamma-interferon mRNA was mainly induced in the CD8+ lymphocyte subset. alpha-Interferon mRNA was expressed largely in the B-cell population, while TNF-alpha mRNA was induced in all leukocyte subsets tested. Therefore, these results suggest that the immunomodulatory effects of FAA in mice are direct, but different cytokines are induced from different leukocyte subsets. Further, the data suggest that flavonoid compounds or analogues that stimulate cytokine gene expression in human cells might be therapeutically active in cancer patients.


Subject(s)
Flavonoids/pharmacology , Interferon-alpha/genetics , Interferon-gamma/genetics , Leukocytes/physiology , Tumor Necrosis Factor-alpha/genetics , Animals , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Immunity, Cellular/drug effects , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/physiology , Mice , RNA, Messenger/genetics , Species Specificity , Spleen/cytology
8.
Biotherapy ; 3(3): 219-32, 1991.
Article in English | MEDLINE | ID: mdl-1906724

ABSTRACT

Lymphokine activated killer (LAK) cells administered in combination with interleukin 2 (IL2) can mediate antitumor activity in tumor-bearing mice and advanced cancer patients. Relatively little is known about the mechanism by which adoptively transferred LAK cells plus IL2 mediate these antitumor effects in vivo, and it remains unclear to what extent the actual LAK effector cells can accumulate in tumors. In the present study, enriched cytolytic LAK effector cells were obtained by fractionation of bulk LAK cell cultures on Percoll density gradients. About 95% of the total lytic activity was recovered from the 55% of cells isolated in fraction 2 (Fr2). The cells recovered in Fr2 are mostly large, proliferating lymphoblasts that express either the NK-associated surface markers NK1.1 (38%) or LGL-1 (31%), or the cytotoxic T cell phenotype, Lyt2 (39%). The cytolytic lymphoblasts obtained from Fr2 were radiolabelled with either 111Indium-Oxine (111InOx) which labels all cells in the population, or with 125Iododeoxyuridine (125IUdR) which labels only proliferating cells, and injected iv into mice bearing murine renal cancer (Renca). 111InOx-labeled Fr2 cells migrated mostly to spleen (28%) and liver (35%), with approximately 5% of the injected label detectable in the Renca-bearing kidney by 24 hrs. In contrast, Fr2 cells labeled with 125IUdR, which labels only the proliferating blasts thought to include the actual effector cells, exhibited a very different localization pattern. 125IUdR-Fr2 cells were retained in the lungs at higher levels than were 111InOx-Fr2 cells and very little label was detectable in liver (6%), spleen (3%), or tumor bearing kidney (2%) at 24 hrs. These results suggest that most of the large, proliferating lymphoblasts are cleared from the body by 24 hrs and very few localize into even large tumors. Subsequently, Northern blot analyses performed on bulk LAK cells revealed a potent induction of mRNA for TNF alpha by 6 hrs and for IFN gamma by 48 hrs. The intensity of gene expression for both cytokines was increased in Fr2 as compared to the unfractionated bulk LAK cells or to non-cytolytic cells obtained from Fr3. Overall, these results suggest that at least some of the antitumor effects mediated by LAK cells occur by the release of cytokines that synergize with exogenous IL2 for the activation of host effector cells.


Subject(s)
Cytokines/biosynthesis , Gene Expression , Killer Cells, Lymphokine-Activated/metabolism , Tumor Cells, Cultured/metabolism , Animals , Blotting, Northern , Cytokines/genetics , Cytotoxicity Tests, Immunologic , Idoxuridine/metabolism , Indium Radioisotopes , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Killer Cells, Lymphokine-Activated/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , RNA, Messenger/metabolism , Tumor Cells, Cultured/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
9.
J Immunol ; 143(1): 372-8, 1989 Jul 01.
Article in English | MEDLINE | ID: mdl-2732472

ABSTRACT

A variety of biologic response modifiers (BRM) can potently augment NK activity in nonlymphoid organs. By using the liver as a model organ, we have shown that this augmentation of organ-associated NK activity is coincident with a 10- to 15-fold increase in the number of large granular lymphocytes (LGL) which can be isolated. The present study was designed to investigate the mechanism by which BRM induce this increase in liver-associated LGL and the coincident increase in hepatic NK activity. Initial studies confirm that a single dose of the pyran copolymer, maleic anhydride divinyl ether (MVE-2), augmented hepatic NK activity and increased the number of liver-associated LGL from 3 x 10(4)/liver to 5 x 10(5)/liver (a 17-fold increase). Multiple injections of MVE-2 further augmented total liver-associated NK activity and LGL number (to 13 x 10(5)/liver). As expected, both the NK activity and detectable LGL were eliminated by treatment of the mice with antiasialo GM1 (asGM1) serum. Three possible mechanisms for the BRM-induced increase in liver-associated LGL have been investigated, including 1) the rapid proliferation of resident hepatic LGL, 2) the redistribution of mature LGL from peripheral sites such as the spleen, or by 3) a rapid output and subsequent hepatic localization of LGL or their precursors recently derived from the bone marrow (BM). Our results demonstrated that the contribution of in situ proliferation to the BRM-induced increase in liver-LGL was relatively small, since the number of cells expressing NK-associated markers (i.e., asGM1, Thy-1.2, and NK1.1) and in G2/M phase (as assessed by propidium iodide uptake) was only 4 to 8%. Further experiments demonstrated that splenectomy before the administration of MVE-2 did not inhibit the augmentation of liver-associated NK activity. This result argued against a recruitment of mature LGL from the spleen. In contrast, selective depletion of the BM following administration of 89Sr decreased the ability of MVE-2 to augment liver-associated NK activity by greater than 80%. This procedure also significantly decreased the ability of Propionibacterium acnes (85%) and multiple doses of IL-2 (49%) to augment liver-associated NK activity. These results demonstrate that the rapid augmentation of liver-associated NK activity by BRM is largely due to localization and accumulation in the liver of LGL recently derived from the BM.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bone Marrow/immunology , Cell Movement , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/immunology , Liver/immunology , Animals , Bone Marrow/drug effects , Bone Marrow/radiation effects , Cell Movement/drug effects , Cell Movement/radiation effects , Cytotoxicity, Immunologic/radiation effects , Drug Administration Schedule , Female , Killer Cells, Natural/drug effects , Killer Cells, Natural/radiation effects , Liver/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/radiation effects , Mice , Mice, Inbred C57BL , Premedication , Pyran Copolymer/administration & dosage , Spleen/radiation effects , Splenectomy , Strontium Radioisotopes/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...