Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Oncogene ; 32(33): 3829-39, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-22964639

ABSTRACT

We report on the role of hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) as an inhibitor of metastasis. HEXIM1 expression is decreased in human metastatic breast cancers when compared with matched primary breast tumors. Similarly we observed decreased expression of HEXIM1 in lung metastasis when compared with primary mammary tumors in a mouse model of metastatic breast cancer, the polyoma middle T antigen (PyMT) transgenic mouse. Re-expression of HEXIM1 (through transgene expression or localized delivery of a small molecule inducer of HEXIM1 expression, hexamethylene-bis-acetamide) in PyMT mice resulted in inhibition of metastasis to the lung. Our present studies indicate that HEXIM1 downregulation of HIF(-)1α protein allows not only for inhibition of vascular endothelial growth factor-regulated angiogenesis, but also for inhibition of compensatory pro-angiogenic pathways and recruitment of bone marrow-derived cells (BMDCs). Another novel finding is that HEXIM1 inhibits cell migration and invasion that can be partly attributed to decreased membrane localization of the 67 kDa laminin receptor, 67LR, and inhibition of the functional interaction of 67LR with laminin. Thus, HEXIM1 re-expression in breast cancer has therapeutic advantages by simultaneously targeting more than one pathway involved in angiogenesis and metastasis. Our results also support the potential for HEXIM1 to indirectly act on multiple cell types to suppress metastatic cancer.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Female , Flow Cytometry , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Immunoprecipitation , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , MCF-7 Cells , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Invasiveness/pathology , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tandem Mass Spectrometry , Tissue Array Analysis , Two-Hybrid System Techniques
3.
J Cancer Res Clin Oncol ; 127(10): 619-24, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11599798

ABSTRACT

PURPOSE: Some limitations of effective therapy in multiple myeloma include the low growth fraction of the malignant plasma cells, multi-drug resistance, and the presence of other concurrent diseases in this patient population. A phase I study was conducted to evaluate the toxicity of granulocyte macrophage colony stimulating factor (GM-CSF) in myeloma patients as well as the potential effect on the plasma cell labeling index (PCLI). Relapsed patients with multiple myeloma were eligible. METHODS: The first phase of this trial assessed the toxicity (including the effect on disease progression) of escalating doses (125-500 microg/m2 SC, days 1-5) of GM-CSF, and the effects of this cytokine on PCLI. Patients whose PCLI doubled and increased to > or = 1.7% were treated with chemotherapy including cyclophosphamide, vincristine, prednisone, and GM-CSF. Twenty-two patients were enrolled. RESULTS: The toxicity of GM-CSF was mild, and no dose-limiting side effects were seen. Twenty-five percent of patients (5/20) achieved the target PCLI, and 4/5 proceeded to receive chemotherapy. No relationship of GM-CSF dose to increases of the PCLI was noted. All patients who received chemotherapy responded. CONCLUSIONS: GM-CSF has acceptable toxicity in patients with multiple myeloma and produced increases of PCLI in selected individuals. Further studies of GM-CSF alone or in combination with chemotherapy are indicated.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Injections, Intravenous/adverse effects , Multiple Myeloma/drug therapy , Alanine Transaminase/blood , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aspartate Aminotransferases/blood , Female , Humans , Male , Multiple Myeloma/pathology , Neoplasm Staging , Patient Selection , Recombinant Proteins
4.
Clin Cancer Res ; 7(3 Suppl): 940s-946s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300495

ABSTRACT

Antitumor immunity fails to adequately develop in many cancer patients, including those with renal cell carcinoma (RCC). A number of different mechanisms have been proposed to explain the immune dysfunction observed in cancer patient T cells. Here we show that T cells from RCC patients display increased sensitivity to apoptosis. Tumor-infiltrating lymphocytes (TILs) display the most profound sensitivity, because 10-15% of those cells are apoptotic when assessed by terminal deoxynucleotidyltransferase-mediated nick end labeling in situ, and the number of apoptotic TILs further increases after 24 h of culture. Peripheral blood T cells from RCC patients are not directly apoptotic, although T lymphocytes derived from 40% of those individuals undergo activation-induced cell death (AICD) upon in vitro stimulation with phorbol myristate acetate and ionomycin. This is in contrast to T cells from normal individuals, which are resistant to AICD. TILs and peripheral blood T cells from RCC patients also exhibit impaired activation of the transcription factor, nuclear factor (NF)-kappaB. Additional findings presented here indicate that the heightened sensitivity of patient T cells to apoptosis may be tumor induced, because supernatants from RCC explants sensitize, and in some instances directly induce, normal T cells to apoptosis. These same supernatants also inhibit NF-kappaB activation. RCC-derived gangliosides may represent one soluble tumor product capable of sensitizing T cells to apoptosis. Pretreatment with neuraminidase, but not proteinase K, abrogated the suppressive effects of tumor supernatants on both NF-kappaB activation and apoptosis. Additionally, gangliosides isolated from tumor supernatants not only inhibited NF-kappaB activation but also sensitized T cells to AICD. These findings demonstrate that tumor-derived soluble products, including gangliosides, may contribute to the immune dysfunction of T cells by altering their sensitivity to apoptosis.


Subject(s)
Apoptosis , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , NF-kappa B/physiology , Cell Nucleus/metabolism , DNA Fragmentation , Enzyme Activation , Gangliosides/metabolism , Humans , In Situ Nick-End Labeling , Ionomycin/pharmacology , Ionophores/pharmacology , Tetradecanoylphorbol Acetate , Time Factors
5.
Cancer J Sci Am ; 6 Suppl 1: S81-7, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685665

ABSTRACT

PURPOSE: The development of an effective antitumor immune response is compromised in patients with renal cell carcinoma. Despite significant infiltration by T lymphocytes into renal tumors, no detectable induction of gene expression is associated with the generation of an antitumor immune response. Tumor-induced down-regulation of interleukin (IL)-2 expression may contribute to the impaired development of the T cell-mediated antitumor immune response. Within renal tumors, there is no detectable expression of IL-2 or the IL-2 receptor alpha chain, and only low levels of interferon gamma (IFN-gamma) mRNA are detected. Products in the tumor environment may suppress the expression of these genes, thus inhibiting production of type 1 helper T cell cytokines. METHODS: Peripheral blood lymphocytes obtained from healthy volunteers were exposed to supernatants from renal cell carcinoma explants, and the immunologic consequences of this were assessed using a variety of molecular assays. RESULTS: Soluble products from renal tumor explants can inhibit the production of IL-2 and IFN-gamma by peripheral blood lymphocytes and can suppress T-cell proliferation. Soluble products from renal cell carcinoma explants appear to block the nuclear translocation of nuclear factor kappa B (NFkappaB) proteins p50 and RelA without affecting cytoplasmic levels of these proteins. In some experiments, a reduction in the nuclear translocation of other transcription factors involved in IL-2 gene expression, including nuclear factor of activated T cells and accessory protein-1, was observed. Gangliosides isolated from tumor supernatants blocked the production of IL-2 and IFN-gamma in response to ionomycin plus phorbol myristate acetate stimulation. These gangliosides also inhibited stimulus-dependent activation and nuclear accumulation of NFkappaB. Coculture experiments demonstrated that renal cell carcinoma lines known to express gangliosides could inhibit the activation of NFkappaB in normal T cells and the Jurkat T-cell line. Supernatants from renal cell carcinoma explants and renal cell carcinoma cell lines can also suppress the proliferation of normal T cells, thus reproducing another defect observed in tumor-infiltrating lymphocytes. Supernatants from renal cell carcinoma tumors also appear to inhibit signaling through the IL-2 receptor. Although tumor supernatants had little effect on IL-2 receptor (alpha, beta or gamma) expression, they did block expression of JAK3, a key kinase involved in signaling through the IL-2 receptor pathway. Moreover, downstream events in IL-2 receptor signaling linked to JAK3 were impaired in T cells treated with tumor supernatants. CONCLUSION: These findings suggest that soluble products from renal tumors may suppress T-cell responses by blocking both IL-2 production and normal IL-2 receptor signaling.


Subject(s)
Carcinoma, Renal Cell/immunology , Interleukin-2/biosynthesis , Kidney Neoplasms/immunology , Receptors, Interleukin-2/physiology , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation , NF-kappa B/metabolism , T-Lymphocytes/immunology , Tumor Cells, Cultured
6.
Clin Cancer Res ; 5(10): 2780-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10537342

ABSTRACT

Interleukin 12 (IL-12) is known to play an important role in the development of an antitumor response. Its activity has been shown to be dependent upon the intermediate production of IFN-gamma and the influx into the tumor of CD8 lymphocytes. In a murine model, tumor regression induced by IL-12 treatment correlated with IFN-gamma, IP-10, and Mig expression in the tumor bed and was abrogated by antibodies to both chemokines. Here we examined the effects of rHuIL-12 on IFN-gamma and CXC chemokine gene expression in patients with renal cell carcinoma (RCC) in an attempt to determine whether a similar series of molecular events leading to IL-12-mediated tumor regression in mice is also detectable in humans. As in the murine RENCA model, cultured RCC cells themselves could be induced by IFN-gamma to synthesize IP-10 and Mig mRNA. Explanted RCC produced IFN-gamma and IP-10 mRNA in response to IL-12 treatment, which was consistent with the finding that biopsied RCC tumors from IL-12-treated patients also variably expressed augmented levels of those molecules after therapy. Although Mig mRNA was present in the majority of biopsied tumors prior to treatment, both the Mig and IP-10 chemokines as well as IFN-gamma were induced in the peripheral blood mononuclear cells of IL-12-treated patients. Skin biopsies of IL-12-treated patients also all synthesized IP-10 mRNA. This study demonstrates that recombinant human IL-12 therapy of patients with RCC has the potential to induce the expression of gene products within the tumor bed that may contribute to the development of a successful antitumor response.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Chemokines, CXC/biosynthesis , Intercellular Signaling Peptides and Proteins , Interferon-gamma/biosynthesis , Interleukin-12/pharmacology , Kidney Neoplasms/drug therapy , Animals , Chemokine CXCL10 , Chemokine CXCL9 , Chemokines, CXC/genetics , Humans , Mice , RNA, Messenger/analysis , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
7.
J Clin Invest ; 104(6): 769-76, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10491412

ABSTRACT

Activation of the transcription factor nuclear factor-kappaB (NFkappaB) is impaired in T cells from patients with renal cell carcinomas (RCCs). In circulating T cells from a subset of patients with RCCs, the suppression of NFkappaB binding activity is downstream from the stimulus-induced degradation of the cytoplasmic factor IkappaBalpha. Tumor-derived soluble products from cultured RCC explants inhibit NFkappaB activity in T cells from healthy volunteers, despite a normal level of stimulus-induced IkappaBalpha degradation in these cells. The inhibitory agent has several features characteristic of a ganglioside, including sensitivity to neuraminidase but not protease treatment; hydrophobicity; and molecular weight less than 3 kDa. Indeed, we detected gangliosides in supernatants from RCC explants and not from adjacent normal kidney tissue. Gangliosides prepared from RCC supernatants, as well as the purified bovine gangliosides G(m1) and G(d1a), suppressed NFkappaB binding activity in T cells and reduced expression of the cytokines IL-2 and IFN-gamma. Taken together, our findings suggest that tumor-derived gangliosides may blunt antitumor immune responses in patients with RCCs.


Subject(s)
Carcinoma, Renal Cell/immunology , Gangliosides/pharmacology , I-kappa B Proteins , Immunosuppressive Agents/pharmacology , Kidney Neoplasms/immunology , NF-kappa B/antagonists & inhibitors , T-Lymphocytes/drug effects , DNA-Binding Proteins/metabolism , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , T-Lymphocytes/metabolism
8.
J Immunol ; 163(2): 590-8, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10395645

ABSTRACT

NF-kappa B is involved in the transcriptional control of various genes that act as extrinsic and intrinsic survival factors for T cells. Our findings show that suppression of NF-kappa B activity with cell-permeable SN50 peptide, which masks the nuclear localization sequence of NF-kappa B1 dimers and prevents their nuclear localization, induces apoptosis in resting normal human PBL. Inhibition of NF-kappa B resulted in the externalization of phosphatidylserine, induction of DNA breaks, and morphological changes consistent with apoptosis. DNA fragmentation was efficiently blocked by the caspase inhibitor Z-VAD-fmk and partially blocked by Ac-DEVD-fmk, suggesting that SN50-mediated apoptosis is caspase-dependent. Interestingly, apoptosis induced by NF-kappa B suppression, in contrast to that induced by TPEN (N,N,N',N'-tetrakis [2-pyridylmethyl]ethylenediamine) or soluble Fas ligand (CD95), was observed in the absence of active death effector proteases caspase-1-like (IL-1 converting enzyme), caspase-3-like (CPP32/Yama/apopain), and caspase-6-like and without cleavage of caspase-3 substrates poly(ADP-ribose) polymerase and DNA fragmentation factor-45. These findings suggest either low level of activation is required or that different caspases are involved. Preactivation of T cells resulting in NF-kappa B nuclear translocation protected cells from SN50-induced apoptosis. Our findings demonstrate an essential role of NF-kappa B in survival of naive PBL.


Subject(s)
Apoptosis/immunology , Caspase 1/metabolism , Caspases/metabolism , Caspases/physiology , NF-kappa B/antagonists & inhibitors , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Apoptosis/drug effects , Caspase 3 , Caspase 6 , Cell Survival/drug effects , Cell Survival/immunology , Enzyme Activation/immunology , Humans , Immunosuppressive Agents/pharmacology , Interphase/immunology , Lymphocyte Activation/drug effects , NF-kappa B/metabolism , NF-kappa B/physiology , Peptides/physiology , Phosphatidylserines/metabolism , Protein Binding/drug effects , Protein Binding/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
9.
Clin Cancer Res ; 5(5): 1219-29, 1999 May.
Article in English | MEDLINE | ID: mdl-10353760

ABSTRACT

Tumors may escape immune recognition and destruction through the induction of apoptosis in activated T lymphocytes. Results from several laboratories suggest that FasL (L/CD95L) expression in tumors may be responsible for this process. In this study of patients with renal cell carcinoma (RCC), we provide evidence for two mechanisms of T-cell apoptosis. One mechanism involves the induction of apoptosis via FasL expression in tumor cells. This is supported by several observations, including the fact that tumor cells in situ as well as cultured cell lines expressed FasL mRNA and protein by a variety of techniques. The FasL in RCC is functional because in coculture experiments, FasL+ tumors induced apoptosis in Fas-sensitive Jurkat T cells and in activated peripheral blood T cells but not in resting peripheral blood T cells. Most importantly, antibody to FasL partially blocked apoptosis of the activated T cells. Moreover, Fas was expressed by T cells derived from the peripheral blood (53% median) and tumor (44.3% median) of RCC patients. Finally, in situ staining for DNA breaks demonstrated apoptosis in a subset of T cells infiltrating renal tumors. These studies also identified a second mechanism of apoptosis in RCC patient peripheral T cells. Whereas these cells did not display DNA breaks when freshly isolated or after culture for 24 h in medium, peripheral blood T cells from RCC patients underwent activation-induced cell death after stimulation with either phorbol 12-myristate 13-acetate/ionomycin or anti-CD3/CD28 antibodies. Apoptosis mediated by exposure to FasL in tumor cells or through T-cell activation may contribute to the failure of RCC patients to develop an effective T-cell-mediated antitumor response.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/cytology , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , T-Lymphocytes, Cytotoxic/cytology , Apoptosis/drug effects , Blood Cells/immunology , Carcinoma, Renal Cell/blood , DNA Fragmentation , Fas Ligand Protein , Humans , In Situ Nick-End Labeling , Ionomycin/pharmacology , Jurkat Cells/immunology , Kidney Neoplasms/blood , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Muromonab-CD3/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , fas Receptor/physiology
11.
Blood ; 93(7): 2308-18, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10090941

ABSTRACT

The Janus kinase, JAK3 plays an important role in interleukin-2 (IL-2)-dependent signal transduction and proliferation of T lymphocytes. Our findings show that prostaglandin E2 (PGE2) can inhibit upregulation of JAK3 protein in naive T cells and can downregulate its expression in primed cells. Reduction in JAK3 was selective because expression of other tyrosine kinases (JAK1, p56(lck), and p59(fyn)) and signal transducer and activator of transcription (STAT)5, which are linked to IL-2 receptor (IL-2R) signaling pathway, were not affected. Inhibition of JAK3 may be controlled by intracellular cyclic adenosine monophosphate (cAMP) levels, as forskolin, a direct activator of adenylate cyclase and dibutyryl cAMP (dbcAMP), a membrane permeable analogue of cAMP suppressed JAK3 expression. Moreover, 3-isobutyl-1-methylxanthine (IBMX), an inhibitor of cAMP phosphodiesterase, potentiated PGE2-induced suppression of JAK3. In naive T cells, but not primed T cells, PGE2 and other cAMP elevating agents also caused a modest reduction in surface expression of the common gamma chain (gammac) that associates with JAK3. The absence of JAK3, but not IL-2R in T cells correlated with impaired IL-2-dependent signal transduction and proliferation. The alteration in IL-2 signaling included decreased tyrosine phosphorylation and DNA binding activity of STAT5 and poor induction of the c-Myc and c-Jun pathways. In contrast, IL-2-dependent induction of Bcl-2 was unaffected. These findings suggest that suppression of JAK3 levels may represent one mechanism by which PGE2 and other cAMP elevating agents can inhibit T-cell proliferation.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Bucladesine/pharmacology , Colforsin/pharmacology , Cyclic AMP/physiology , Dinoprostone/pharmacology , Protein-Tyrosine Kinases/biosynthesis , Receptors, Interleukin-2/physiology , Second Messenger Systems/drug effects , T-Lymphocytes/drug effects , Adenylyl Cyclases/metabolism , Enzyme Induction/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-2/pharmacology , Janus Kinase 3 , Phorbol 12,13-Dibutyrate/pharmacology , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/pharmacology , Second Messenger Systems/physiology , T-Lymphocytes/enzymology
12.
J Immunother ; 22(1): 71-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9924702

ABSTRACT

Interleukin-12 (IL-12) is a heterodimeric cytokine that enhances the cytolytic activity, proliferation, and interferon-gamma (IFN-gamma) production by T lymphocytes and natural killer cells, and has significant antitumor activity in a variety of murine tumor models. The induction of interferon (IFN)-gamma by IL-12 in tumor-bearing mice plays an important role in its antitumor activity. We therefore examined the effects of IL-12 on IFN-gamma production by T cells derived from patients with renal cell carcinoma (RCC), including freshly isolated tumor infiltrating lymphocytes (T-TIL), matched peripheral blood T cells (T-PBL), and RCC-specific TIL lines. IL-12 alone induced IFN-gamma secretion by T cells from normal individuals and appeared to act synergistically with either IL-2 or anti-CD3 antibody. In contrast, it failed to stimulate significant IFN-gamma secretion by T-PBL and T-TIL from RCC patients. This unresponsive state in T-PBL appeared selective because IFN-gamma was produced when cells were stimulated with either phytohemagglutinin or anti-CD3 antibody. Moreover, costimulation through the T-cell receptor (TCR)/CD3 complex or with IL-2 made T-PBL from RCC patients responsive to IL-12, possibly secondary to the upregulation of IL-12R (beta chain). A selective loss of IL-12-dependent production of IFN-gamma was also consistently observed in two of three established RCC-specific TIL lines. Although these cell lines did not respond to any concentration of IL-12, they did produce IFN-gamma after ligation of the TCR/CD3 or stimulation with IL-2, IL-12 also acted either syngeristically or additively with IL-2, anti-CD3 antibody, or autologous tumor cells to induce IFN-gamma production. The observed decreases in IFN-gamma production in response to IL-12 may have a negative effect on the development of T-cell immunity. The clinical importance of these findings during in vivo administration of IL-12 remains to be determined.


Subject(s)
Carcinoma, Renal Cell/immunology , Interferon-gamma/biosynthesis , Interleukin-12/pharmacology , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Carcinoma, Renal Cell/drug therapy , Cell Separation , DNA/biosynthesis , Flow Cytometry , Humans , Interleukin-2/pharmacology , Kidney Neoplasms/drug therapy , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Muromonab-CD3/immunology , Phytohemagglutinins/pharmacology , Receptors, Interleukin/metabolism , Receptors, Interleukin-12 , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects , Tumor Cells, Cultured
13.
Clin Cancer Res ; 4(10): 2337-47, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9796963

ABSTRACT

Studies have demonstrated abnormalities of the CD3/T-cell antigen receptor (TCR) and pathways of signal transduction in T lymphocytes from animals and patients with advanced malignancy. Diminished expression of TCRzeta and p56(lck) that are associated with the TCR and reduced nuclear localization of RelA containing nuclear factor kappaB (NFkappaB) complexes have been noted. These defects have been described in T cells from patients with malignant melanoma, renal cell carcinoma (RCC), ovarian cancer, and colorectal cancer. Preliminary observations also indicate possible correlation with clinical variables such as stage in selected instances. To further characterize altered expression of TCRzeta, p56(lck), and impaired activation of NFkappaB, T lymphocytes were obtained from 65 patients with RCC, the majority of whom were receiving combination cytokine therapy [interleukin (IL)-2, IFN alpha-containing regimens] and 37 control individuals. In 29 of these patients, levels of TCRzeta and p56(lck) were determined by Western blots of T-cell lysates and semiquantitated using densitometry. Relative levels were then correlated with a series of clinical variables including response to therapy, performance status, survival, disease sites, age, and others. In another group of 28 patients (three individuals from the first group), the frequency of abnormal NFkappaB activation was studied using electrophoretic mobility shift assays after activation of T cells with phorbol myristate acetate/ionomycin or anti-CD3 monoclonal antibody. Changes in these signaling molecules during cytokine treatment were also investigated. TCRzeta and p56(lck) were detected in the peripheral blood T cells in 27 of 29 patients, and overall, reduced levels were noted visually in 12 of 29 (41%) and 13 of 29 (45%) individuals, respectively. When levels were semiquantitated using densitometry, significant decreases of TCRzeta (P = 0.029) and p56(lck) (P = 0.029) but not CD3epsilon (P = 0.131), compared with control levels, were found. In patients treated with IL-2/IFN alpha-based therapy, relative levels of TCRzeta increased significantly (P = 0.002) on day 15 of cycle one compared with the baseline. Correlations of TCRzeta or p56(lck) levels with response or disease variables, except for lower TCRzeta levels (P < 0.001) in the presence of bone metastases, were not found. Abnormal NFkappaB activation after stimulation with phorbol myristate acetate/ionomycin and/or anti-CD3 monoclonal antibody was found in 59% of patients (17 of 28) and was not accounted for by the advanced age of the study cohort. Activation of NFkappaB in peripheral blood T cells was inducible during cytokine therapy in four of six individuals who displayed impaired NFkappaB activity prior to therapy. Moreover, impaired activation of NFkappaB does not appear linked to a reduction of TCRzeta expression, because in five patients, normal TCRzeta levels were present although kappaB binding was not inducible. In the majority of patients with advanced RCC, peripheral blood T cells express TCRzeta and p56(lck), and in a subset, reduced levels of these TCRzeta associated molecules are seen that may increase during cytokine-based therapy. Abnormal activation of NFkappaB is also present in >50% of patients and may also revert to normal during IL-2/IFN alpha-based treatment. This alteration in NFkappaB activation occurred in the presence of normal expression of TCRzeta-associated signaling elements. The clinical significance of these findings remains unclear.


Subject(s)
Carcinoma, Renal Cell/immunology , Cytokines/therapeutic use , Kidney Neoplasms/immunology , Signal Transduction , T-Lymphocytes/metabolism , Carcinoma, Renal Cell/therapy , Humans , Kidney Neoplasms/therapy , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/analysis , Membrane Proteins/analysis , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/analysis
14.
Blood ; 92(4): 1334-41, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9694722

ABSTRACT

Activation of the transcription factor NFkappaB in peripheral blood T cells from patients with renal cell carcinoma (RCC) is compromised. This impaired signaling function results from a failure of RelA and c-Rel to translocate to the nucleus though normal levels of Rel proteins are present in the cytoplasm. We demonstrate here in a subset of RCC patients that the defect in NFkappaB activation is attributable to the absence of phosphorylation and degradation of the inhibitor IkappaBalpha. In patient T cells there was no stimulus dependent decrease in the cytoplasmic level of IkappaBalpha. Coimmunoprecipitation studies showed that RelA was in complex with IkappaBalpha and was not released after stimulation. Moreover, the phosphorylated form of IkappaBalpha detected in normal T cells after activation is absent in patient T cells. Additional experiments showed that soluble products from RCCs (RCC-S) can reproduce the same phenotype in T cells from healthy individuals. Supernatant fluid from cultured explants of RCC, but not normal kidney, inhibited the stimulus dependent nuclear translocation of NFkappaB without altering the cytoplasmic levels of RelA, c-Rel, and NFkappaB1. Phosphorylation and degradation of IkappaBalpha was also blocked by RCC-S. The mechanistic similarities between patient-derived T cells and normal T cells cultured with RCC-S suggest that the tumor-derived products may be the primary mediators of impaired T-cell function in this tumor system.


Subject(s)
Carcinoma, Renal Cell/immunology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/immunology , NF-kappa B/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Transcription Factors , Biological Transport , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Nucleus/metabolism , Culture Media, Conditioned/pharmacology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-rel , Signal Transduction , Transcription Factor RelA , Transcription Factor RelB , Transcription, Genetic
15.
J Immunother ; 20(6): 479-87, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409454

ABSTRACT

Granzyme B is a protein thought to play a pivotal role in the cytolytic functions of T cells. In view of this, the inducibility of this gene in freshly isolated T cells (T-TILs) infiltrating human renal cell carcinoma (RCC) in vitro was examined by using the reverse transcriptase-polymerase chain reaction (RT-PCR). A reduction in granzyme B messenger RNA (mRNA) expression in stimulated T-TILs from five of nine patients with RCC compared with autologous peripheral blood T cells was noted. The reduced expression was observed after multiple stimuli including anti-CD3 antibody, interleukin-2 (IL-2), and phytohemagglutinin (PHA). Because CD8+ T cells represent the predominant cytotoxic population, the ability of this cell population to express granzyme B mRNA after stimulation also was examined. When compared with CD8+ peripheral blood lymphocytes (T-PBLs) from patients with RCC and normal donors, the induction of granzyme B mRNA was reduced in CD8+ T-TILs. CD8+ T-TILs also had lower non-major histocompatibility complex (MHC)-restricted cytotoxic activity than did CD8+ T-PBLs against both Daudi cells and allogeneic RCC cell lines. These results show that in a subset of patients with RCC, depressed lytic activity of CD8+ TILs compared with CD8+ PBLs is present. Reduced granzyme B mRNA expression also was noted in selected patients.


Subject(s)
Carcinoma, Renal Cell/immunology , Gene Expression , Kidney Neoplasms/immunology , Serine Endopeptidases/genetics , T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , Granzymes , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA-Directed DNA Polymerase , Serine Endopeptidases/metabolism , T-Lymphocytes/immunology
16.
J Immunol ; 159(6): 3057-67, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9300731

ABSTRACT

The proliferative capacity of T cells infiltrating human tumors is known to be impaired, possibly through their interaction with tumor. Here we demonstrate that soluble products derived from renal cell carcinoma (RCC-S) explants but not normal kidney can inhibit an IL-2-dependent signaling pathway that is critical to T cell proliferation. A major target of the immunosuppression was the IL-2R-associated protein tyrosine kinase, Janus kinase 3 (Jak3). RCC-S suppressed basal expression of Jak3 and its increase following stimulation with anti-CD3/IL-2. Jak3 was most sensitive to suppression by RCC-S; however, reduction in expression of p56(lck), p59(fyn), and ZAP-70 was observed in some experiments. Expression of other signaling elements linked to the IL-2R (Jak1) and the TCR (TCR-zeta, CD3-epsilon, and phospholipase C-gamma) were minimally affected. In naive T cells, RCC-S also partially blocked induction of IL-2R alpha-, beta- and gamma-chain expression when stimulating via the TCR/CD3 complex with anti-CD3 Ab. To determine whether RCC-S suppressed IL-2-dependent signaling, primed T cells were employed since RCC-S had no effect on IL-2R expression but did down-regulate Jak3 expression and, to a lesser degree, p56(lck) and p59(fyn). Reduction in Jak3 correlated with impaired IL-2-dependent proliferation and signal transduction. This included loss of Jak1 kinase tyrosine phosphorylation and no induction of the proto-oncogene, c-Myc. These findings suggest that soluble products from tumors may suppress T cell proliferation through a mechanism that involves down-regulation of Jak3 expression and inhibition of IL-2-dependent signaling pathways.


Subject(s)
Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Protein-Tyrosine Kinases/immunology , Receptors, Interleukin-2/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Tumor Escape , Cell Division/immunology , Cytotoxicity, Immunologic , Humans , Janus Kinase 3 , Lymphocyte Activation/immunology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Mas , Receptors, Interleukin-2/antagonists & inhibitors , Tumor Cells, Cultured
17.
J Immunother ; 20(4): 301-11, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9220320

ABSTRACT

A phase II trial was conducted in patients with metastatic renal cell carcinoma, to assess the clinical efficacy and immunoregulatory effects of continuous-infusion recombinant interleukin-2 (rIL-2) (9.0 x 10(6) IU/m2/day on days 1-5, 8-12, 15-19, and 22-26) and subcutaneously administered recombinant human interferon-alpha 2b (rHuIFN alpha 2b) (10.0 x 10(6) U/m2/day TIW). Thirty-six patients with metastatic renal cell carcinoma, performance status of 0-1, and measurable disease who had not received prior rIL-2, rHuIFN alpha 2b, or chemotherapy were treated. Patients with CNS metastases, active infections, history of another malignancy within 3 years, and those requiring corticosteroids were ineligible. Cycles of rIL-2 and rHuIFN alpha 2b were administered in the outpatient department every 6-8 weeks in stable or responding patients until patient tolerance or a complete response were reached. Doses were modified for grade III or IV toxicity. Ancillary studies included three-color immunocytometric analysis of peripheral blood lymphocytes, repetitive tumor biopsies for immunohistologic analysis of infiltrating cells and proliferative responses of tumor infiltrating lymphocytes, and preliminary studies of changes in peripheral blood T-lymphocyte signal transduction molecules [T-cell receptor (TCR)-zeta, p56ick, p59fyn]. Thirty-six eligible patients were treated, with 6 of 36 patients (17%, 95% confidence interval 6-33%) responding (3 complete response, 3 partial response). In two of the partial responders, and in an additional three patients with either minimal tumor regression (one patient) or stable disease (two patients), surgical removal of residual disease was undertaken. The median survival of all patients was 14 months. The toxicity of this regimen was severe, but outpatient administration was possible in most instances. Immunoregulatory effects on T-cell subsets included increases in various CD3+ CD25+/- HLADr+/- subsets unrelated to response. Tumor biopsies before and/or during therapy were obtained in 17 patients, and no consistent alterations in the degree of T-lymphocyte or macrophage infiltrates could be detected. In a subset of patients, tumor infiltrating lymphocyte proliferative responses and levels of peripheral blood T-cell signal transduction molecules (TCR-zeta, p56lck, p59fyn) were investigated. Abnormalities were found in selected patients, which improved during rIL-2/rHuIFN alpha 2b therapy. This cytokine combination produces tumor regression in selected patients with metastatic renal cell carcinoma. Surrogate immunologic markers associated with response were not identified; however, preliminary studies demonstrate investigation of immune defects and their reversal with cytokine therapy is possible.


Subject(s)
Carcinoma, Renal Cell/therapy , Interferon-alpha/administration & dosage , Interleukin-2/administration & dosage , Kidney Neoplasms/therapy , Adult , Aged , Antigens, CD/analysis , Carcinoma, Renal Cell/immunology , Female , Humans , Interferon alpha-2 , Interferon-alpha/adverse effects , Interleukin-2/adverse effects , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Recombinant Proteins
18.
Clin Cancer Res ; 2(2): 347-57, 1996 Feb.
Article in English | MEDLINE | ID: mdl-9816178

ABSTRACT

We conducted a Phase I trial of s.c. recombinant human interleukin 3 (rhIL-3) to evaluate the toxicity, maximal tolerated dose, pharmacokinetics, and in vivo biological effects of this cytokine. Thirty-one patients with refractory cancer were entered into the study between November 1991 and June 1993. Therapy consisted of s.c. rhIL-3 daily for 15 days administered to cohorts of three to nine patients at dose levels of 60-4000 microgram/m2/day. Cycles were repeated at intervals of 28 days. Seventy-five cycles of rhIL-3 were administered (median, two per patient) and the maximal tolerated dose was 2000 microgram/m2/day. Toxicity was moderate, with most patients developing chills, fever, and myalgia. Dose-limiting toxicity consisted of diarrhea (two patients) and headache (one patient). Hematological effects of rhIL-3 included significant dose-related increases of WBC (P < 0.001), neutrophils (P < 0.001), and eosinophils (P < 0.001). Platelet counts and absolute lymphocyte numbers also increased. Various CD3(+) lymphocyte subsets increased; however, lytic activity (natural killer and lymphokine-activated killer) of peripheral blood lymphocytes was not enhanced. Serum levels of the soluble IL-2 receptor increased in a dose-related fashion, and IL-2-induced lymphocyte proliferation also was increased variably. Pharmacokinetic studies were performed in 13 patients, and area under the curve and maximal concentration values increased with increasing rhIL-3 dose levels (P < 0.001) and correlated with maximal changes from baseline in WBC, neutrophils, and eosinophils. rhIL-3 antibodies were detected in 8% of patients by day 29 of cycle 1 but were not neutralizing. rhIL-3 is well tolerated when administered s.c. and has reproducible hematological and immunological effects. The pleiotropic effects of this cytokine on various in vivo biological parameters were demonstrated clearly. Further studies of its immunoregulatory effects are warranted.


Subject(s)
Interleukin-3/adverse effects , Neoplasms/therapy , Adult , Aged , Blood Cell Count/drug effects , Female , Humans , Injections, Subcutaneous , Interleukin-3/administration & dosage , Interleukin-3/pharmacokinetics , Interleukin-6/blood , Lymphocyte Activation/drug effects , Male , Middle Aged , Neoplasms/blood , Neoplasms/immunology , Receptors, Interleukin-2/analysis , Recombinant Proteins/adverse effects
19.
Cancer Immunol Immunother ; 41(3): 175-84, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7553687

ABSTRACT

T cells infiltrating (T-TIL) B cell non-Hodgkin's lymphomas (NHL) are thought to represent a local host response to the tumor. However, tumor progression in the presence of this T cell infiltrate suggests that the T-TIL may be functionally impaired. To address this issue we determined whether response to stimulation of T-TIL from 25 patients with NHL through the T cell receptor (TCR/CD3) and the interleukin-2 (IL-2) receptor (IL-2R) was intact, since activation of these receptors is important for proliferation and cytokine production. Our results demonstrate defects in response to stimulation via TCR/CD3 and the IL-2R in T-TIL cells from patients with NHL that were not observed with T cells from the peripheral blood. T-TIL showed minimal proliferation to anti-CD3 and only modest proliferation to IL-2 alone or when combined with anti-CD3. Moreover, cytokine production in T-TIL was impaired since stimulation through the TCR/CD3 complex did not induce mRNA for interferon gamma (IFN gamma), IL-2, IL-4 or IL-10. The functional unresponsiveness of these cells may be linked to altered signalling through the TCR/CD3 since an abnormal tyrosine phosphorylation pattern was detected in T-TIL after stimulation with anti-CD3.


Subject(s)
Lymphocyte Activation/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/immunology , Receptors, Antigen, T-Cell/physiology , Receptors, Interleukin-2/physiology , Adult , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Base Sequence , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-2/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/physiology , Lymphoma, B-Cell/metabolism , Molecular Sequence Data , Phosphorylation , Tyrosine/metabolism
20.
Cancer Res ; 54(20): 5424-9, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7923175

ABSTRACT

Recent data suggest that the poor induction of a T-cell response to human renal cell carcinoma (RCC) may be related to alterations in signal transduction pathways. We report that T cells from RCC patients have two alterations in kappa B motif-specific DNA-binding activity. The first alteration involves the constitutive expression of substantial kappa B-binding activity in nuclear extracts, which was observed in the electrophoretic mobility shift assay. The magnitude of kappa B activity in unstimulated patient T cells was similar to that observed in T cells from normal individuals that had been activated in vitro. On the basis of Western blotting experiments using antibodies to kappa B/Rel family proteins, the kappa B-binding activity constitutively expressed in T cells from RCC patients is composed mostly of the NF-kappa B1 (p50) subunit. The second abnormality in kappa B-binding activity in T cells from these patients is that RelA, a member of the Rel homology family which is part of the normal NF-kappa B complex, was not induced in the nucleus following activation. Western blotting analysis did not detect any RelA in nuclear extracts either before or after stimulation of T cells. The altered kappa B-binding activity in T cells from RCC patients may impair their capacity to respond normally to various stimuli.


Subject(s)
Carcinoma, Renal Cell/immunology , DNA, Neoplasm/metabolism , Kidney Neoplasms/immunology , NF-kappa B/metabolism , T-Lymphocytes/immunology , Base Sequence , Blotting, Western , Humans , Molecular Sequence Data , Signal Transduction , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...