Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Br J Cancer ; 108(2): 319-26, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23322198

ABSTRACT

BACKGROUND: Sunitinib (Su), a tyrosine kinase inhibitor of VEGFR, is effective at producing tumour response in clear cell renal cell carcinoma (cRCC), but resistance to therapy is inevitable. As COX-2 is a known mediator of tumour growth, we explored the potential benefit of COX-2 inhibition in combination with VEGFR inhibition in attempts at delaying tumour progression on Su. METHODS: COX-2 expression was compared with areas of hypoxia in tumours that progressed on Su vs untreated tumours. Mice bearing human cRCC xenografts were treated with Su and the COX-2 inhibitor, celecoxib, and the effects on tumour growth were assessed. Sequential vs concurrent regimens were compared. RESULTS: COX-2 expression was increased in cRCC xenografts in areas of tumour hypoxia. The combination of Su and celecoxib achieved longer times to tumour progression compared to treatment with either agent alone or to untreated control animals in four models. This effect was seen with concurrent but not with sequential therapy. CONCLUSION: COX-2 inhibition can extend the effectiveness of VEGFR inhibition. This effect is dependent on the timing of therapy. Clinical trials combining Su and COX-2 inhibitors should be considered as a means delaying time to progression on sunitinib in patients with metastatic cRCC.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Cyclooxygenase 2/metabolism , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyrroles/therapeutic use , Sulfonamides/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Celecoxib , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Female , Humans , Indoles/administration & dosage , Indoles/pharmacology , Mice , Pyrazoles/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sulfonamides/pharmacology , Sunitinib , Xenograft Model Antitumor Assays
3.
Cell Growth Differ ; 12(11): 543-50, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714635

ABSTRACT

The antibiotic radicicol suppresses transformation in a variety of transformed cells. The antineoplastic effects of the drug have been attributed to the degradation of Raf and the inactivation of the Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling cascade. Here we demonstrate that radicicol induces cell spreading, suppresses anchorage-independent cell growth, and increases the expression of the high-molecular weight tropomyosin isoform TM-2 in cells stably expressing a constitutively active form of MEK-1 as well as in ras-transformed cells. Furthermore, the reverting effects of the drug are achieved at concentrations below those required to deplete Raf from the cell or to inhibit the phosphorylation of ERK or its substrates Elk and pp90(RSK). In contrast, low concentrations of radicicol significantly inhibited activator protein (AP-1) and serum response factor (SRF)-mediated transcription. The lack of correlation between the effects of radicicol on cell phenotype and on the signaling activities of the Raf/MEK/ERK pathway indicate that Raf depletion or disruption of proximal signaling events in the mitogen-activated protein kinase pathway are not the predominant mechanisms by which the drug suppresses the transformed phenotype. Our observation that low concentrations of radicicol block transcriptional activities mediated by AP-1 and SRF suggests that interference with signaling upstream of these transcription factors may contribute to the reverting effects of the drug.


Subject(s)
Cell Transformation, Neoplastic/drug effects , DNA-Binding Proteins , Lactones/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Oncogene Protein p21(ras)/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Transcription Factors , Tropomyosin/metabolism , 3T3 Cells , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Line, Transformed , Cell Size/drug effects , Dose-Response Relationship, Drug , Fibroblasts , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Kinase 1 , Macrolides , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation, Missense/genetics , Oncogene Protein p21(ras)/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Tropomyosin/genetics , ets-Domain Protein Elk-1
4.
Blood ; 98(9): 2720-5, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11675343

ABSTRACT

Baseline platelet production is dependent on thrombopoietin (TPO). TPO is constitutively produced and primarily regulated by receptor-mediated uptake by platelets. Inflammatory thrombocytosis is thought to be related to increased interleukin-6 (IL-6) levels. To address whether IL-6 might act through TPO to increase platelet counts, TPO was neutralized in vivo in C57BL/10 mice treated with IL-6, and hepatic TPO mRNA expression and TPO plasma levels were studied. Transcriptional regulation of TPO mRNA was studied in the hepatoblastoma cell line HepG2. Furthermore, TPO plasma levels were determined in IL-6-treated cancer patients. It is shown that IL-6-induced thrombocytosis in C57BL/10 mice is accompanied by enhanced hepatic TPO mRNA expression and elevated TPO plasma levels. Administration of IL-6 to cancer patients results in a corresponding increase in TPO plasma levels. IL-6 enhances TPO mRNA transcription in HepG2 cells. IL-6-induced thrombocytosis can be abrogated by neutralization of TPO, suggesting that IL-6 induces thrombocytosis through TPO. A novel pathway of TPO regulation by the inflammatory mediator IL-6 is proposed, indicating that the number of platelets by themselves might not be the sole determinant of circulating TPO levels and thus of thrombopoiesis. This regulatory pathway might be of relevance for the understanding of reactive thrombocytosis.


Subject(s)
Blood Platelets/cytology , Hematopoiesis/drug effects , Interleukin-6/pharmacology , Thrombopoietin/drug effects , Animals , Humans , Inflammation/chemically induced , Inflammation/etiology , Interleukin-6/physiology , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Platelet Count , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Thrombocytosis/chemically induced , Thrombocytosis/etiology , Thrombopoietin/blood , Thrombopoietin/genetics , Thrombopoietin/pharmacology , Tumor Cells, Cultured
5.
J Immunother ; 24(1): 91-8, 2001.
Article in English | MEDLINE | ID: mdl-11211153

ABSTRACT

Interleukin-12 (IL-12) is a cytokine with effects on immune function and hematopoiesis. In this article, the authors describe two patients with renal cell cancer in whom grade 4 neutropenia and grade 3 hemolytic anemia developed, respectively, during treatment with twice-weekly intravenous recombinant human interleukin-12 (rhIL-12) during a phase 1 trial. The severe neutropenia was associated with bone marrow agranulocytosis and a preponderance of large granular lymphocytes in the peripheral blood, whereas the hemolytic anemia was negative for the Coombs test and associated with splenomegaly. The agranulocytosis and hemolytic anemia persisted after the rhIL-12 was stopped, but both subsequently responded to treatment with cyclophosphamide. steroids, or both. These findings indicate that rhIL-12 can induce unique hematologic toxic effects that can be reversed with immunosuppressive drugs.


Subject(s)
Anemia, Hemolytic/immunology , Carcinoma, Renal Cell/drug therapy , Interleukin-12/adverse effects , Interleukin-12/therapeutic use , Kidney Neoplasms/drug therapy , Neutropenia/immunology , Aged , Anemia, Hemolytic/chemically induced , Female , Humans , Injections, Intravenous , Interleukin-12/administration & dosage , Male , Middle Aged , Neutropenia/chemically induced , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use
6.
J Biol Chem ; 276(10): 6893-6, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11145953

ABSTRACT

The caspase-8 homologue FLICE-inhibitory protein (FLIP) functions as a caspase-8 dominant negative, blocking apoptosis induced by the oligomerization of the adapter protein FADD/MORT-1. FLIP expression correlates with resistance to apoptosis induced by various members of the tumor necrosis factor family such as TRAIL. Furthermore, forced expression of FLIP renders cells resistant to Fas-mediated apoptosis. Although FLIP expression is regulated primarily by MEK1 activity in activated T cells, the oncogenic signaling pathways that regulate FLIP expression in tumor cells are largely unknown. In this report, we examined the roles of the MAP kinase and phosphatidylinositol (PI) 3-kinase signaling pathways in the regulation of FLIP expression in tumor cells. We observed that the MEK1 inhibitor PD98059 reduced FLIP levels in only 2 of 11 tumor cell lines tested. In contrast, disruption of the PI 3-kinase pathway with the specific inhibitor LY294002 reduced Akt (protein kinase B) phosphorylation and the levels of FLIP protein and mRNA in all cell lines evaluated. The introduction of a dominant negative Akt adenoviral construct also consistently reduced FLIP expression as well as the phosphorylation of the Akt target glycogen synthase kinase-3. In addition, infection of the same cell lines with a constitutively active Akt adenovirus increased FLIP expression and the phosphorylation of GSK-3. These data add FLIP to the growing list of apoptosis inhibitors in which expression or function is regulated by the PI 3-kinase-Akt pathway.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis , Apoptosis Regulatory Proteins , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, Dominant , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , MAP Kinase Kinase 1 , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Signal Transduction , T-Lymphocytes/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
7.
J Immunol ; 165(7): 4120-6, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11034424

ABSTRACT

IL-12 plays a pivotal role in the stimulation of immune responses against intracellular infections. This role is manifested in the increased susceptibility to atypical mycobacterial and salmonella infections among individuals whose lymphocytes lack expression of IL-12Rbeta1. Here, we report on a patient with Mycobacterium avium infection, recurrent Staphylococcus aureus sinusitis, and multiple adverse drug reactions whose T cells were unable to produce IFN-gamma or proliferate in response to IL-12 despite the expression of wild-type IL-12Rbeta1 and IL-12Rbeta2. The defect in these functional responses to IL-12 was selective, as cytolytic activity induced by IL-12 was intact, and lymphocytes were responsive to stimulation by IL-2. An examination of cytokine signaling revealed that STAT4 and extracellular regulated kinase 1 (ERK1) activation by IL-12 was intact, whereas the activation of STAT1, -3, and -5 by IL-12 was lost. This impairment of STAT activation was specific for IL-12, as STAT activation by IL-2, IL-15, and IFN-gamma was unaffected. These findings demonstrate that the activation of STAT4 alone is not sufficient for IL-12-induced IFN-gamma production and proliferation and suggest that other STATs play a role in these responses to IL-12. While the etiology of the impaired IL-12 signaling in this patient has not yet been elucidated, the absence of mutations in IL-12Rbeta1 or IL-12Rbeta2 and the preservation of STAT4 activation raise the possibility that there may be a mutation in an as yet undiscovered component of the IL-12 signaling complex that is normally required for the recruitment and activation of STAT1, -3, and -5.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-12/physiology , Milk Proteins , Mycobacterium avium-intracellulare Infection/immunology , Signal Transduction/immunology , Staphylococcal Infections/immunology , Trans-Activators/metabolism , Child, Preschool , Cytotoxicity, Immunologic , DNA-Binding Proteins/deficiency , Enzyme Activation/immunology , Female , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/microbiology , Infant , Interferon-gamma/biosynthesis , Interferon-gamma/deficiency , Interleukin-12/metabolism , Lymphocyte Activation , Mitogen-Activated Protein Kinases/metabolism , Mycobacterium avium-intracellulare Infection/metabolism , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/chemistry , Receptors, Interleukin/genetics , Receptors, Interleukin-12 , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT4 Transcription Factor , STAT5 Transcription Factor , Staphylococcal Infections/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , Trans-Activators/deficiency
8.
Clin Cancer Res ; 6(6): 2201-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10873069

ABSTRACT

In an effort to develop a biochemotherapy regimen for metastatic melanoma suitable for testing in a cooperative group setting, we modified the concurrent biochemotherapy regimen of S. S. Legha et al. (J. Clin. Oncol., 16: 1752-1759, 1998) by providing enhanced supportive care and developing a strict, conservative approach to the management of treatment-related toxicities. Patients received cisplatin, vinblastine, and dacarbazine (CVD: cisplatin (20 mg/m2) and vinblastine (1.2 mg/m2) on days 1-4, dacarbazine (800 mg/m2) on day 1 only) concurrently with interleukin 2 (9 MIU/m2/day) by continuous i.v. infusion on days 1-4 and IFN-alpha (5 MU/m2/day) on days 1-5, 8, 10, and 12. Prophylactic antibiotics and a maximum of four cycles were administered. Routine granulocyte colony-stimulating factor and aggressive antiemetics were initiated after patients 7 and 14, respectively. Forty-four patients were enrolled in this study. No patients had received prior chemotherapy or interleukin 2; however, 23 (53%) had received prior IFN-alpha, mostly in the adjuvant setting. A total of 131 treatment cycles was administered. Significant toxicities requiring dose modification included: hypotension requiring pressors (15 episodes in 11 patients), grades 3/4 vomiting (12 episodes in 15 cycles; 5 episodes in 12 patients (6 episodes in 9 cycles after initiation of the modified antiemetic regimen), transient renal insufficiency (5 episodes in 5 patients), grade 4 thrombocytopenia (24 episodes, 1 associated with bleeding), neutropenia with or without fever (15 instances, only 11 in 112 cycles after routine use of granulocyte colony-stimulating factor), and catheter-related bacteremia (2 patients). Five (16%) of 30 patients who were treated after the last protocol modification experienced what we defined as unacceptable toxicity for a cooperative group setting. Responses were seen in 19 of 40 evaluable patients (relative risk, 48%) with 8 complete responses (20%). The median response duration was 7 months (range, 1-17+ months) with one currently ongoing. The central nervous system was the initial site of relapse in 11 responding patients. The median survival duration was 11 months (range, 2-31 months). This modified, concurrent biochemotherapy regimen is active and tolerable for use in a cooperative group setting. Central nervous system relapse, however, remains a concern for responders. This regimen is being compared with CVD in a Phase III Intergroup Trial (Eastern Cooperative Oncology Group/Southwest Oncology Group 3695).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Neoplasms/secondary , Cisplatin/administration & dosage , Cisplatin/adverse effects , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Disease-Free Survival , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Male , Middle Aged , Neoplasm Metastasis , Pilot Projects , Recombinant Proteins , Recurrence , Time Factors , Treatment Outcome , Vinblastine/administration & dosage , Vinblastine/adverse effects
9.
Clin Cancer Res ; 6(5): 1678-92, 2000 May.
Article in English | MEDLINE | ID: mdl-10815886

ABSTRACT

The aim of this study was to examine the tolerability, antitumor activity, and biological effects of a new schedule of i.v. recombinant human interleukin 12 (rhIL-12). Twenty-eight patients were enrolled in a Phase I trial in which rhIL-12 was administered twice weekly as an i.v. bolus for 6 weeks. Stable or responding patients were eligible to receive additional 6-week cycles until there was no evidence of disease or until tumor progression. Patient cohorts were treated with escalating doses of rhIL-12 (30-700 ng/kg). The maximum tolerated dose (MTD) was 500 ng/kg, with dose-limiting toxicities consisting of elevated hepatic transaminases and cytopenias. At the MTD (n = 14), there was one partial response occurring after 6 cycles of rhIL-12 in a patient with renal cell cancer. Two additional renal cell cancer patients treated at the MTD had prolonged disease stabilization, with one of these exhibiting tumor regression after 8 cycles of rhIL-12. IFN-gamma, IL-15, and IL-18 were induced in patients treated with rhIL-12. Whereas IFN-gamma and IL-15 induction were attenuated midway through the first cycle in patients with disease progression, those patients with tumor regression or prolonged disease stabilization were able to maintain IFN-gamma, IL-15, and IL-18 induction. The down-modulation of IFN-gamma induction during rhIL-12 treatment did not relate to IL-10 production or alterations in rhIL-12 bioavailability but was associated with an acquired defect in lymphocyte IFN-gamma production in response to IL-12, IL-2, or IL-15. This defect could be partially overcome in vitro through combined stimulation with IL-12 plus IL-2. These findings show that the chronic administration of twice-weekly i.v. rhIL-12 is well-tolerated, stimulates the production of IL-12 costimulatory cytokines and IFN-gamma, and can induce delayed tumor regression. Strategies aimed at maintaining IFN-gamma induction, such as the addition of IL-2, may further augment the response rate to this schedule of rhIL-12.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Interleukin-12/therapeutic use , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Adult , Aged , Arthralgia/chemically induced , Carcinoma, Renal Cell/pathology , Dose-Response Relationship, Drug , Female , Fever/chemically induced , Hematologic Diseases/chemically induced , Humans , Hypotension/chemically induced , Injections, Intravenous , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-12/adverse effects , Interleukin-12/pharmacokinetics , Interleukin-15/metabolism , Interleukin-18/metabolism , Kidney Neoplasms/pathology , Male , Melanoma/pathology , Middle Aged , Mouth Mucosa , Neoplasm Metastasis , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Skin/drug effects , Skin/pathology , Stomatitis/chemically induced , Time Factors
10.
J Biol Chem ; 275(2): 1209-15, 2000 Jan 14.
Article in English | MEDLINE | ID: mdl-10625665

ABSTRACT

We isolated and identified an endogenous 24-kDa human basement membrane-derived inhibitor of angiogenesis and tumor growth, termed canstatin. Canstatin, a fragment of the alpha2 chain of type IV collagen, was produced as a recombinant molecule in Escherichia coli and 293 embryonic kidneys cells. Canstatin significantly inhibited human endothelial cell migration and murine endothelial cell tube formation. Additionally, canstatin potently inhibited 10% fetal bovine serum-stimulated endothelial cell proliferation and induced apoptosis, with no inhibition of proliferation or apoptosis observed on non-endothelial cells. Inhibition of endothelial proliferation was not concomitant with a change in extracellular signal-regulated kinase activation. We demonstrate that apoptosis induced by canstatin was associated with a down-regulation of the anti-apoptotic protein, FLIP. Canstatin also suppressed in vivo growth of large and small size tumors in two human xenograft mouse models with histology revealing decreased CD31-positive vasculature. Collectively, these results suggest that canstatin is a powerful therapeutic molecule for suppressing angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Collagen/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Neovascularization, Physiologic/drug effects , Animals , Apoptosis/drug effects , Cattle , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Cells, Cultured , Cloning, Molecular/methods , Collagen/biosynthesis , Collagen/chemistry , Collagen/genetics , Endothelium, Vascular/drug effects , Escherichia coli , Humans , Mice , Pulmonary Artery , Recombinant Proteins/pharmacology , Transfection
11.
Int J Immunopathol Pharmacol ; 13(1): 1-12, 2000.
Article in English | MEDLINE | ID: mdl-12749772

ABSTRACT

L-selectin is down-modulated from the cell surface in response to leukocyte activation or cross-linking with ligand mimetics such as anti-L-selectin antibodies or sulfatides. The down-modulation induced upon cellular activation with PMA is due to proteolytic shedding mediated by a cysteine metalloprotease, presumably the TNF-alpha converting enzyme (TALE), and is susceptible to inhibitors of the hydroxamate class. To determine if cross-linking-induced down-modulation of L-selectin is similarly affected by these inhibitors, we exposed Jurkat T cells to the anti-L-selectin antibody Dreg 200 or sulfatides in the presence of the hydroxamate TNF-alpha protease inhibitor (TAPI). TAPI completely inhibited PMA-induced downmodulation but had no effect on that induced by the anti-L-selectin antibody or sulfatides. The downmodulation induced by an anti-TCR antibody (WT31) was only partially inhibited by TAPI. An L-selectin mutant lacking the putative TACE cleavage site and resistant to PMA-induced shedding (321del.9) was expressed in L1.2 pre-B cells. Like WT L-selectin, this mutant was down-modulated when exposed to sulfatides, or Dreg 200 and this down-modulation was unaffected by TAPI. An L-selectin cytoplasmic tail deletion mutant (344del.15) expressed in L1.2 pre-B cells was down-modulated by PMA or sulfatides, but not Dreg 200. Electrophoretic analysis of L-selectin immunoprecipitated from the supernatants of Jurkat cells treated with either sulfatides or D200 revealed a proteolytic fragment of the same size as that released from the cell surface in response to PMA. Our data indicate that the down regulation of L-selectin in response to cross-linking by ligands or TCR engagement may be mechanistically distinct from that induced by PMA. Furthermore, our results with the 344del.15 mutant suggest that the down-modulation of Lselectin induced by certain sulfated carbohydrate ligands may be initiated through surface receptors other than L-selectin itself. The abbreviations used in this paper are: TAPI, TNF-alpha protease inhibitor; TACE, TNF-alpha converting enzyme; PMA, 4(3-phorbol 12-myristate 13-acetate.

12.
Eur Cytokine Netw ; 10(1): 37-42, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10210771

ABSTRACT

Interleukin-1 (IL-1) is a critical mediator of inflammation. Two naturally occurring IL-1 antagonists have been described, namely the IL-1 receptor antagonist (IL-1Ra) and the IL-1 receptor type II (IL-1RII). IL-1RII does not transmit a signal upon binding of IL-1, but competes with the signaling of IL-1RI for binding of IL-1. Shedding of IL-1RII yields the soluble IL-1 receptor type II (IL-1sRII) which retains the ability of membrane-bound IL-1RII to bind IL-1beta avidly, but binds IL-1Ra and IL-1alpha with low affinity. In contrast, IL-1sRI retains the ability of membrane-bound IL-1RI to bind IL-1Ra and IL-1alpha with high affinity, but binds IL-1beta poorly. We have previously shown that immunotherapy with IL-2 or IL-6 in cancer patients is associated with a dramatic increase in IL-1Ra plasma levels. In the present study, plasma levels of soluble IL-1 receptors were monitored in healthy individuals and cancer patients. In healthy controls, the mean IL-1sRII level was 4.76 0.16 ng/ml. IL-1sRII levels in cancer patients were comparable to those measured in healthy controls. IL-1sRII levels did not vary during the first 52 hours after initiation of IL-2 therapy, but increased significantly thereafter to reach 9.56 1.16 ng/ml on day 5. In contrast, IL-6 immunotherapy with a 5-day continuous infusion did not trigger an increase in IL-1sRII levels. IL-1sRI levels did not increase during immunotherapy with IL-2 or IL-6. Our results indicate that IL-1sRII, unlike IL-1Ra, remains a modest, natural, anti-inflammatory mechanism triggered by immunotherapy with IL-2, but not with IL-6.


Subject(s)
Carcinoma, Renal Cell/immunology , Interleukin-2/therapeutic use , Interleukin-6/therapeutic use , Kidney Neoplasms/immunology , Melanoma/immunology , Receptors, Interleukin-1/blood , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Humans , Immunotherapy , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Melanoma/blood , Melanoma/pathology , Melanoma/therapy , Receptors, Interleukin-1 Type II , Reference Values , Time Factors
13.
J Biol Chem ; 273(48): 32182-6, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9822696

ABSTRACT

Transformation is accompanied by the down-regulation of the high molecular weight isoforms of non-muscle tropomyosin. Several lines of evidence suggest that tropomyosin down-regulation may be essential for ras-induced tumorigenicity. It is unclear which of the many signaling pathways downstream of Ras are involved in tropomyosin down-regulation. Here we demonstrate that Raf activation induces tropomyosin down-regulation comparable to that induced by Ras. Expression of the effector-domain mutant Ras-G12V,Y40C, which is unable to bind Raf, induced only modest down-modulation of tropomyosin. Treatment with the MEK-specific inhibitor PD98059 had little effect on tropomyosin levels in ras- or raf-transformed cells. In contrast, a mutant form of MEK-1, MEK-1-S218A,S222A, restored tropomyosin levels in ras-transformed NIH3T3 cells almost to the levels observed in non-transformed cells. MEK-1-S218A,S222A does not inhibit MEK phosphorylation and is a poor inhibitor of ERK phosphorylation. These data suggest that this mutant form of MEK-1 interferes with a yet uncharacterized pathway controlled by Raf. We conclude that the ras-induced down-modulation of tropomyosin is predominantly Raf-mediated, but MEK-independent, and that a novel pathway exists downstream of Raf which may play an important role in regulation of the cytoskeleton.


Subject(s)
Genes, ras , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Retroviridae Proteins, Oncogenic/genetics , Tropomyosin/genetics , 3T3 Cells , Amino Acid Substitution , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line, Transformed , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , MAP Kinase Kinase 1 , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Oncogene Proteins v-raf , Oncogenes , Phosphorylation , Point Mutation , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Retroviridae Proteins, Oncogenic/biosynthesis , Transfection , Tropomyosin/biosynthesis
14.
Clin Cancer Res ; 4(5): 1203-13, 1998 May.
Article in English | MEDLINE | ID: mdl-9607578

ABSTRACT

Our purpose was to determine the maximum tolerated dose and toxicity associated with soluble Chinese hamster ovary [s(CHO)] recombinant human interleukin (IL) 1 receptor (IL-1R; Immunex, Seattle, WA) administration in humans and to determine the effective biological dose and/or maximum tolerated dose of the s(CHO) IL-1R in combination with high-dose IL-2 as determined by reduction in IL-2 toxicity and modulation of its biological effects. Twenty-seven patients with metastatic cancer were treated with escalating doses of s(CHO) IL-1R at 1, 1, 5, 10, 20, 40, and 55 mg/m2 i.v. on days -6 (except cohort 2), 1, and 15 and IL-2 at doses of 300,000 IU/kg (cohort 1) and 600,000 IU/kg (cohorts 2-7) i.v. every 8 h on days 1-5 and 15-19. No toxicity directly attributable to s(CHO) IL-1R was observed. The median number of IL-2 doses was 23. Hypotension and neurotoxicity were the major dose-limiting toxicities for the IL-2/s(CHO) IL-1R combination. Of the 24 patients treated with full-dose IL-2, there were six responses, three complete and three partial (response rate, 25%). Three patients developed thyroid dysfunction, and all 3 responding melanoma patients exhibited vitiligo. The t1/2 of s(CHO) IL-1R alone was 24-30 h and was not significantly altered by coadministration with IL-2. Whole-blood functional assays indicated that sufficient s(CHO) IL-1R was present in the circulation at top dose levels to inhibit the in vitro effects of IL-1beta on IL-8 induction; however, no effect on IL-2-induced IL-8 induction, or on the IL-1beta- or IL-2-induced tumor necrosis factor production, was observed. Suppression of IL-2-mediated tumor necrosis factor alpha and IL-6 induction in vivo during the first 24 h after IL-2 administration was observed, and the neutrophil chemotactic defect normally seen with IL-2 was not observed. IL-1R antagonist induction far exceeded that seen previously with IL-2 alone. No inhibition of either serum C-reactive protein induction or enhanced urinary nitrate excretion and no consistent effect on IL-2-related changes in peripheral blood mononuclear cell phenotype or endothelial adhesion molecule expression were seen. The coadministration of s(CHO) IL-1R produced no apparent reduction in IL-2 clinical toxicity manifested by either the ability to administer more IL-2 than anticipated or a reduction in the toxicity associated with a given amount of IL-2. Therefore, no effective biological dose could be identified for the s(CHO) IL-1R.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Renal Cell/drug therapy , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Receptors, Interleukin-1/administration & dosage , Adult , Aged , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Autoimmunity/drug effects , C-Reactive Protein/drug effects , C-Reactive Protein/metabolism , CHO Cells/metabolism , Carcinoma, Renal Cell/metabolism , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cricetinae , Cytokines/drug effects , Cytokines/metabolism , Drug Administration Schedule , Female , Humans , Immunophenotyping , Interleukin-2/adverse effects , Interleukin-2/pharmacokinetics , Kidney Neoplasms/metabolism , Male , Melanoma/metabolism , Middle Aged , Neutrophils/drug effects , Neutrophils/physiology , Nitrates/urine , Receptors, Interleukin-1/metabolism
15.
J Cell Biol ; 141(3): 647-62, 1998 May 04.
Article in English | MEDLINE | ID: mdl-9566966

ABSTRACT

Previous studies suggest that the Ca2+-dependent proteases, calpains, participate in remodeling of the actin cytoskeleton during wound healing and are active during cell migration. To directly test the role that calpains play in cell spreading, several NIH-3T3- derived clonal cell lines were isolated that overexpress the biological inhibitor of calpains, calpastatin. These cells stably overexpress calpastatin two- to eightfold relative to controls and differ from both parental and control cell lines in morphology, spreading, cytoskeletal structure, and biochemical characteristics. Morphologic characteristics of the mutant cells include failure to extend lamellipodia, as well as abnormal filopodia, extensions, and retractions. Whereas wild-type cells extend lamellae within 30 min after plating, all of the calpastatin-overexpressing cell lines fail to spread and assemble actin-rich processes. The cells genetically altered to overexpress calpastatin display decreased calpain activity as measured in situ or in vitro. The ERM protein ezrin, but not radixin or moesin, is markedly increased due to calpain inhibition. To confirm that inhibition of calpain activity is related to the defect in spreading, pharmacological inhibitors of calpain were also analyzed. The cell permeant inhibitors calpeptin and MDL 28, 170 cause immediate inhibition of spreading. Failure of the intimately related processes of filopodia formation and lamellar extension indicate that calpain is intimately involved in actin remodeling and cell spreading.


Subject(s)
Actins/metabolism , Calpain/metabolism , 3T3 Cells , Animals , Calcium-Binding Proteins/genetics , Calpain/antagonists & inhibitors , Cell Movement , Cysteine Proteinase Inhibitors/pharmacology , Cytoskeletal Proteins , Down-Regulation , Humans , Mice , Phosphoproteins/metabolism , Substrate Specificity , Up-Regulation
16.
Immunol Today ; 18(9): 428-32, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9293158

ABSTRACT

Acute inflammation is accompanied by changes in the concentrations of acute phase proteins (APPs). While much is known about the cytokines involved in the initiation of inflammation, less is known about the mediators involved in its resolution. Recent data suggest that interleukin 6 (IL-6) and IL-6-regulated APPs are anti-inflammatory and immuno-suppressive, and may negatively regulate the acute phase response.


Subject(s)
Acute-Phase Proteins/immunology , Interleukin-6/immunology , Allergy and Immunology/trends , Animals , Anti-Inflammatory Agents/immunology , Central Nervous System/immunology , Cytokines/biosynthesis , Humans , Immune Tolerance , In Vitro Techniques , Interleukin-6/genetics , Interleukin-6/pharmacology , Mice , Mice, Knockout , Models, Biological
17.
Mol Biol Cell ; 8(5): 897-908, 1997 May.
Article in English | MEDLINE | ID: mdl-9168473

ABSTRACT

The levels of high molecular weight isoforms of tropomyosin (TM) are markedly reduced in ras-transformed cells. Previous studies have demonstrated that the forced expression of tropomyosin-1 (TM-1) induces reversion of the transformed phenotype of ras-transformed fibroblasts. The effects of the related isoform TM-2 on transformation are less clear. To assess the effects of forced expression of the TM-2 protein on ras-induced tumorigenicity, we introduced a TM-2 cDNA lacking the 3' untranslated region riboregulator into ras-transformed NIH 3T3 fibroblasts. TM-2 expression resulted in a flatter cell morphology and restoration of stress fibers. TM-2 expression also significantly reduced growth rates in low serum, soft agar, and nude mice. The reduced growth rates were associated with a prolongation of G0-G1. To identify the mechanism of TM-2-induced growth inhibition, we analyzed the effects of TM-2 reexpression of ERK and c-jun N-terminal kinase (JNK) activities. Levels of ERK phosphorylation and activity in TM-2-transfected tumor cells were comparable to those in mock-transfected tumor cells. JNK activity was only modestly increased in ras-transformed cells relative to untransformed NIH 3T3 cells and only slightly reduced as result of forced TM-2 expression. We conclude that the partially restored expression of the TM-2 protein induces growth inhibition of ras-transformed NIH 3T3 cells without influencing ERK or JNK activities. Furthermore, the 3' untranslated region riboregulator of the alpha-tropomyosin gene is not needed for the inhibition of ras-induced growth.


Subject(s)
Cell Division , Cell Transformation, Neoplastic , Genes, ras , Mitogen-Activated Protein Kinases , Tropomyosin/genetics , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/genetics , Cell Division/physiology , Cell Line, Transformed , DNA, Complementary , Gene Expression Regulation , Introns , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phenotype , Protein-Tyrosine Kinases/metabolism , Tropomyosin/physiology
18.
J Clin Oncol ; 15(3): 1052-62, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9060545

ABSTRACT

PURPOSE: A randomized, double-blind, placebo-controlled trial was performed to compare the toxicity and biologic effects of treatment with high-dose intravenous (IV) bolus interleukin-2 (IL-2) plus the recombinant human soluble p75 tumor necrosis factor (TNF) receptor immunoglobulin G (IgG) chimera (rhuTNFR:Fc) with high-dose IL-2 alone in patients with advanced melanoma and renal cell carcinoma. PATIENTS AND METHODS: Twenty patients with advanced melanoma or renal cell carcinoma were randomized to receive IL-2 (Chiron, Emeryville, CA) 600,000 IU/kg every 8 hours on days 1 to 5 and 15 to 19 (maximum, 28 doses) combined with placebo or the rhuTNFR:Fc fusion protein (Immunex, Seattle, WA) 10 mg/m2 on days 1 and 15 and 5 mg/m2 on days 3, 5, 17, and 19. The impact of rhuTNFR:Fc on IL-2 toxicity and biologic effects was evaluated. RESULTS: No clinically significant difference in toxicity was observed in the two treatment arms. The adjusted median number of IL-2 doses administered during cycle 1 was 24.5 (range, seven to 28) and 21.5 (range, five to 27) for the placebo and rhuTNFR:Fc arms, respectively (P = .544). IL-2-induced TNF bioactivity, neutrophil chemotactic defect, and serum IL-6, IL-8, and IL-1 receptor antagonist (IL-1RA) induction were suppressed by rhuTNFR:Fc. Two of nine assessable patients (22%) on IL-2/placebo and three of 10 patients (30%) on IL-2/rhuTNFR:Fc responded. CONCLUSION: Despite evidence of in vitro neutralization of TNF functional activity and partial inhibition of other secondary biologic effects of IL-2, rhuTNFR:Fc does not reduce the clinical toxicity associated with high-dose IL-2 therapy. These results suggest that the toxicity and antitumor effects of IL-2 treatment are independent of circulating TNF.


Subject(s)
Antigens, CD , Carcinoma, Renal Cell/drug therapy , Immunoglobulin G/therapeutic use , Interleukin-2/therapeutic use , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/therapeutic use , Skin Neoplasms/drug therapy , Adult , Aged , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/pathology , Double-Blind Method , Female , Humans , Immunoglobulin G/adverse effects , Immunoglobulin G/blood , Immunophenotyping , Interleukin-2/adverse effects , Interleukin-2/blood , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Male , Melanoma/blood , Melanoma/pathology , Middle Aged , Monocytes , Receptors, Tumor Necrosis Factor, Type II , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/blood , Skin Neoplasms/blood , Skin Neoplasms/pathology
19.
Clin Cancer Res ; 3(3): 409-17, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815699

ABSTRACT

A Phase I dose escalation trial of i.v. administered recombinant human interleukin 12 (rhIL-12) was performed to determine its toxicity, maximum tolerated dose (MTD), pharmacokinetics, and biological and potential antineoplastic effects. Cohorts of four to six patients with advanced cancer, Karnofsky performance >/=70%, and normal organ function received escalating doses (3-1000 ng/kg/day) of rhIL-12 (Genetics Institute, Inc.) by bolus i.v. injection once as an inpatient and then, after a 2-week rest period, once daily for five days every 3 weeks as an outpatient. Therapy was withheld for grade 3 toxicity (grade 4 hyperbilirubinemia or neutropenia), and dose escalation was halted if three of six patients experienced a dose-limiting toxicity (DLT). After establishment of the MTD, eight more patients were enrolled to further assess the safety, pharmacokinetics, and immunobiology of this dose. Forty patients were enrolled, including 20 with renal cancer, 12 with melanoma, and 5 with colon cancer; 25 patients had received prior systemic therapy. Common toxicities included fever/chills, fatigue, nausea, vomiting, and headache. Fever was first observed at the 3 ng/kg dose level, typically occurred 8-12 h after rhIL-12 administration, and was incompletely suppressed with nonsteroidal anti-inflammatory drugs. Routine laboratory changes included anemia, neutropenia, lymphopenia, hyperglycemia, thrombocytopenia, and hypoalbuminemia. DLTs included oral stomatitis and liver function test abnormalities, predominantly elevated transaminases, which occurred in three of four patients at the 1000 ng/kg dose level. The 500 ng/kg dose level was determined to be the MTD. This dose, administered by this schedule, was associated with asymptomatic hepatic function test abnormalities in three patients and an onstudy death due to Clostridia perfringens septicemia but was otherwise well tolerated by the 14 patients treated in the dose escalation and safety phases. The T1/2 elimination of rhIL-12 was calculated to be 5.3-9.6 h. Biological effects included dose-dependent increases in circulating IFN-gamma, which exhibited attenuation with subsequent cycles. Serum neopterin rose in a reproducible fashion regardless of dose or cycle. Tumor necrosis factor alpha was not detected by ELISA. One of 40 patients developed a low titer antibody to rhIL-12. Lymphopenia was observed at all dose levels, with recovery occurring within several days of completing treatment without rebound lymphocytosis. There was one partial response (renal cell cancer) and one transient complete response (melanoma), both in previously untreated patients. Four additional patients received all proposed treatment without disease progression. rhIL-12 administered according to this schedule is biologically and clinically active at doses tolerable by most patients in an outpatient setting. Nonetheless, additional Phase I studies examining different schedules and the mechanisms of the specific DLTs are indicated before proceeding to Phase II testing.


Subject(s)
Interleukin-12/adverse effects , Neoplasms/therapy , Adult , Aged , Creatinine/blood , Dose-Response Relationship, Drug , Female , Humans , Hyperglycemia/chemically induced , Injections, Intravenous , Interleukin-12/administration & dosage , Interleukin-12/pharmacokinetics , Liver/drug effects , Male , Metabolic Clearance Rate , Middle Aged , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics
20.
J Immunol ; 157(12): 5653-9, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8955218

ABSTRACT

The adhesion molecule L-selectin (CD62L) is rapidly shed from the plasma membrane during leukocyte activation as a result of proteolytic cleavage between Lys321 and Ser322 within the extracellular domain. L-selectin is also down-modulated from the surface in response to cross-linking, possibly through a similar mechanism. To further characterize the mechanism of down-modulation, several L-selectin mutants were generated and transfected into COS cells. Wild-type L-selectin as well as mutants with one or two amino acid substitutions at the cleavage site were nearly quantitatively shed into the culture supernatant. However, mutants in which a nine-amino acid stretch that included the protease-sensitive site was either deleted or replaced with a polyglycine spacer or a comparable region of E-selectin were retained on the cell surface and not detected in the supernatant. These results are consistent with other reports describing protease resistant L-selectin mutants. We also demonstrate that when expressed in L1-2 pre-B cells, the L-selectin nine-amino acid deletion mutant (321del.9), but not wild-type L-selectin, is resistant to down-regulation induced by PMA. However, both wild-type and mutant 321del.9 are completely lost from the cell surface in response to cross-linking with an L-selectin Ab. PMA-induced- but not L-selectin cross-linking-induced down-modulation was inhibited by staurosporine. These data are consistent with the idea that the L-selectin protease(s) can tolerate minor structural alterations at the cleavage site, and that L-selectin down-modulation can be induced by more than one mechanism, at least one of which (cross-linking) is protein kinase C independent.


Subject(s)
L-Selectin/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , Cross-Linking Reagents , Down-Regulation , Endopeptidases/metabolism , Humans , Immunologic Techniques , L-Selectin/genetics , Lymphocyte Activation , Molecular Sequence Data , Mutagenesis, Site-Directed , Structure-Activity Relationship , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL