Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Apoptosis ; 10(1): 219-31, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15711938

ABSTRACT

Apoptosis is characterized by chromatin condensation, DNA cleavage, redistribution of phosphatidylserine, and apoptotic body formation via an actin-dependent process. We describe a novel form of the execution phase of apoptosis in human multiple myeloma cells that is morphologically and mechanistically distinct from classical apoptosis, but is caspase-dependent and inhibited by IL-6 and overexpression of Bcl-2. Electron microscopic analysis of these cells demonstrated chromatin condensation without nuclear fragmentation, and 'partitioning' of cell constituents into two components: a single, large bleb containing soluble protein and free ribosomes, and a region containing the nucleus, organelles, and RER. In some cases, the bleb separated, becoming a free vesicle exhibiting random kinetic motion. These morphologic features occurred despite inhibition of the actin and tubulin cytoskeletal systems. This novel form of apoptosis, called partitioning apoptosis, was observed in a variety of tumor cell types and in primary cells. The execution phase of apoptosis can occur in a manner that is morphologically and mechanistically distinct from classical apoptosis.


Subject(s)
Apoptosis , Caspases/metabolism , Cell Compartmentation , Chromatin/pathology , Multiple Myeloma/pathology , Multiple Myeloma/ultrastructure , Cells, Cultured , Humans , Microscopy, Electron
2.
Bone Marrow Transplant ; 33(8): 781-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14767498

ABSTRACT

High-dose chemotherapy using melphalan (HDMEL) is an important component of many conditioning regimens that are given before autologous hematopoietic stem cell transplantation (AHSCT). In contrast to the situation in myeloma, and to a lesser degree acute leukemia, only a very limited published experience exists with the use of HDMEL conditioning as a single agent in doses requiring AHSCT for lymphoma, both Hodgkin lymphoma (HL) and especially non-Hodgkin lymphoma (NHL). Thus, we report results of treating 26 lymphoma patients (22 with NHL and four with HL) with HDMEL 220-300 mg/m(2) plus amifostine (AF) cytoprotection and AHSCT as part of a phase I-II trial. Median age was 51 years (range 24-62 years); NHL histology was varied, but was aggressive (including transformed from indolent) in 19 patients, indolent in two patients and mantle cell in one. All 26 patients had been extensively treated; 11 were refractory to the immediate prior therapy on protocol entry and two had undergone prior AHSCT. All were deemed ineligible for other, 'first-line' AHSCT regimens. Of these 26 patients, 22 survived to initial tumor evaluation on D +100. At this time, 13 were in complete remission, including four patients who were in second CR before HDMEL+AF+AHSCT. Responses occurred at all HDMEL doses. Currently, seven patients are alive, including five without progression, with a median follow-up in these latter patients of D +1163 (range D +824 to D +1630); one of these patients had a nonmyeloablative allograft as consolidation on D +106. Conversely, 14 patients relapsed or progressed, including five who had previously achieved CR with the AHSCT procedure. Two patients, both with HL, remain alive after progression; one is in CR following salvage radiotherapy. Six patients died due to nonrelapse causes, including two NHL patients who died while in CR. We conclude that HDMEL+AF+AHSCT has significant single-agent activity in relapsed or refractory NHL and HL. This experience may be used as a starting point for subsequent dose escalation of HDMEL (probably with AF) in established combination regimens.


Subject(s)
Amifostine/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Hematopoietic Stem Cell Transplantation , Hodgkin Disease/therapy , Lymphoma, Non-Hodgkin/therapy , Melphalan/administration & dosage , Radiation-Protective Agents/administration & dosage , Adult , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Transplantation Conditioning/methods , Transplantation, Autologous
3.
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
4.
J Immunol ; 163(4): 1943-50, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10438930

ABSTRACT

In this study four murine IL-12 naked DNA expression plasmids (pIL-12), containing both the p35 and p40 subunits, were shown to induce systemic biological effects in vivo after intradermal injection. Three of the four IL-12 expression vectors augmented NK activity and induced expression of the IFN-gamma and IFN-gamma-inducible Mig genes. Both IL-12 p70 heterodimer and IFN-gamma proteins were documented in the serum within 24 h after intradermal injection of the pIL-12o- plasmid, which also induced the highest level of NK activity in the spleen and liver among the IL-12 constructs. Interestingly, both p40 mRNA expression at the injection site and serum protein levels followed a biphasic pattern of expression, with peaks on days 1 and 5. Subsequent studies revealed that the ability of intradermally injected pIL-12o- to augment NK lytic activity was prevented by administration of a neutralizing anti-IL-12 mAb. Finally, injection of the pIL-12o- into BALB/c IL-12 p40-/- mice also resulted in a biphasic pattern of IL-12 p70 appearance in the serum, and induced IFN-gamma protein and activated NK lytic activity in liver and spleen. These results demonstrate that injection of delivered naked DNA encoding the IL-12 gene mediates the biphasic systemic production of IL-12-inducible genes and augments the cytotoxic function of NK cells in lymphoid and parenchymal organs as a direct result of transgene expression. The results also suggest that these naked DNA plasmids may be useful adjuvants for vaccines against infectious and neoplastic diseases.


Subject(s)
DNA, Viral/administration & dosage , Intercellular Signaling Peptides and Proteins , Interleukin-12/biosynthesis , Interleukin-12/genetics , Killer Cells, Natural/immunology , Lymphocyte Activation/genetics , Th1 Cells/metabolism , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Monoclonal/pharmacology , Chemokine CXCL9 , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Cytomegalovirus/genetics , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Female , Gene Expression Regulation, Viral/immunology , Immunosuppressive Agents/pharmacology , Injections, Intradermal , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-12/administration & dosage , Interleukin-12/immunology , Kinetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Spleen/immunology , Spleen/metabolism , beta-Galactosidase/administration & dosage , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
5.
Cancer Gene Ther ; 5(5): 281-91, 1998.
Article in English | MEDLINE | ID: mdl-9824047

ABSTRACT

The ability to specifically and efficiently express selected genes in tumor cells is an important goal for cancer gene therapy. Transcriptional targeting of adenovirus to tumor cells, thereby limiting their expression to specific cell types, represents one experimental approach to this problem. We have previously shown that a recombinant adenovirus containing the murine tyrosinase promoter coupled to a dimer of the tyrosinase-enhancer element can target the expression of beta-galactosidase cDNA to melanoma cells. We now report that this same promoter/enhancer cassette can efficiently drive the expression of the herpes simplex virus thymidine kinase gene in melanoma cells. Infection of melanoma cells with the AdmTyr-tk virus along with subsequent ganciclovir treatment induces S phase cell cycle arrest associated with a profound change in cell size and morphology. Treated cells remain viable for prolonged periods, but clonogenic assays demonstrate that the cell cycle arrest is irreversible. In contrast, nonmelanoma cells are unaffected by this treatment regimen, exhibiting normal growth kinetics, metabolic activity, and cell cycle progression. The therapeutic efficacy of the AdmTyr-tk virus was tested in vivo using a xenograft model of human melanoma. The injection of the AdmTyr-tk virus into established subcutaneous tumor nodules in combination with systemic ganciclovir administration led to a decreased tumor growth rate and to complete tumor regressions in some cases. These studies demonstrate the feasibility of selectively targeting growth-inhibitory genes to melanoma cells in vitro and in vivo.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Melanoma/therapy , Monophenol Monooxygenase/genetics , Thymidine Kinase/genetics , Animals , Carcinogenicity Tests , Cell Cycle/drug effects , Cell Death/drug effects , Cell Death/genetics , Enhancer Elements, Genetic , Ganciclovir/pharmacology , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Neoplasms, Experimental/therapy , Promoter Regions, Genetic , S Phase/drug effects , Simplexvirus/enzymology , Tumor Cells, Cultured
6.
J Immunol ; 161(8): 3957-65, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9780164

ABSTRACT

Mice bearing the experimental murine renal cancer Renca can be successfully treated with some forms of immunotherapy. In the present study, we have investigated the molecular pathways used by NK and T cells to lyse Renca cells. Renca cells normally express low levels of Fas that can be substantially enhanced by either IFN-gamma or TNF-alpha, and the combination of IFN-gamma + TNF-alpha synergistically enhances cell-surface Fas expression. In addition, cells pretreated with IFN-gamma and TNF-alpha are sensitive to lysis mediated by Fas ligand (FasL)-expressing hybridomas (dllS), cross-linking of anti-Fas Abs or soluble Fas (FasL). Lysis via Fas occurs by apoptosis, since Renca shows all the typical characteristics of apoptosis. No changes in levels of bcl-2 were observed after cytokine treatments. We also examined cell-mediated cytotoxic effects using activated NK cells and T cells from gld FasL-deficient mice, and perforin-deficient mice, as well as wild-type C57BL/6 and BALB/c mice. Interestingly, the granule-mediated pathway predominated in killing of Renca by activated NK cells, while the Fas/FasL pathway contributed significantly to cell-mediated killing of Renca by activated T cells. These results suggest that killing of Renca tumor cells by immune effector cells can occur by both granule and Fas-mediated cytotoxicity. However, for the Fas-mediated pathway to function, cell surface levels of Fas need to be increased beyond a critical threshold level by proinflammatory cytokines such as IFN-gamma and TNF-alpha.


Subject(s)
Cytotoxicity, Immunologic , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Membrane Glycoproteins/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , fas Receptor/immunology , Animals , Cell Death/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Perforin , Pore Forming Cytotoxic Proteins , Signal Transduction/immunology , fas Receptor/genetics
7.
Cancer Res ; 58(17): 3855-65, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731495

ABSTRACT

The mechanisms by which cytotoxic agents perturb the normal cell biology and cell cycle progression of cancer cells were explored using B16F10 cells genetically modified to express the Herpes Simplex Virus-thymidine kinase gene. Culture in the presence of the nucleoside analogue ganciclovir induced a profound morphological change that required entry of treated cells into S phase and was dependent on prenylated proteins such as those of the rho gene family. Cell cycle arrest occurred in late S phase or G2 phase due to the activation of the G2-M DNA damage checkpoint. This checkpoint control operated at the level of inhibition of the activity of Cdc2/cyclin B and occurred by two mechanisms: (a) p53-mediated up-regulation of p21CIP/WAF1 expression and its association with Cdc2/cyclin B; and (b) prevention of the dephosphorylation of tyrosine 15 of Cdc2. These events occurred in vitro and in vivo, and were shown to mediate bystander killing in this model. The mechanism of cell death seemed to be due to the irreversible cell cycle arrest at the G2-M checkpoint, rather than induction of apoptosis. These data link DNA damage checkpoints with cytoskeletal signaling pathways and the core cell cycle machinery and may represent a general mechanism of cytotoxicity of this class of nucleoside analogues.


Subject(s)
Antineoplastic Agents/pharmacology , Cytoskeleton/drug effects , G2 Phase/drug effects , Ganciclovir/pharmacology , Mitosis/drug effects , Animals , Cyclin B/antagonists & inhibitors , DNA Damage , Female , Lovastatin/pharmacology , Mice , Mice, Inbred C57BL , Protamine Kinase/antagonists & inhibitors , Tumor Cells, Cultured
8.
Cancer Res ; 58(15): 3391-400, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9699671

ABSTRACT

The ras oncogene plays an important role in the multistep progression to cancer by activation of signal transduction pathways that contribute to aberrant growth regulation. Although many of these effects are cell autonomous, the ras oncogene also regulates the expression of genes that alter host/tumor interactions. We now extend the mechanisms through which ras promotes tumor survival by demonstrating that oncogenic Ras inhibits expression of the fas gene and renders Ras-transformed cells resistant to Fas-induced apoptosis. A panel of Ras-transformed clones exhibited a marked inhibition in fas mRNA and Fas cell surface expression as compared with untransformed parental cell lines. Fas expression was induced by culture in the presence of IFN-gamma + tumor necrosis factor alpha; however, the maximal level attained in Ras transformants was approximately 10-fold below the level of untransformed cells. Whereas untransformed cells were sensitive to apoptotic death induced by cross-linking surface Fas (especially after cytokine treatment), Ras-transformed cells were very resistant to Fas-induced death even under the most stringent assay conditions. To demonstrate that this resistance was mediated by oncogenic Ras and not secondary genetic events, pools of Ras-transformed cells were generated using a highly efficient retroviral transduction technique. Transformed pools were assayed 6 days after infection and demonstrated a marked decrease in fas gene expression and Fas-mediated apoptosis. Oncogenic Ras did not promote general resistance to apoptosis, because ectopic expression of a fas cDNA in Ras-transformed cells restored sensitivity to Fas-induced apoptosis. These data indicate that oncogenic Ras inhibits basal levels of expression of the fas gene, and although cytokine signal transduction pathways are functional in these cells, the level of surface Fas expression remains below the threshold required for induction of apoptosis. These data identify a mechanism by which Ras-transformed cells may escape from host-mediated immune destruction.


Subject(s)
Apoptosis/physiology , Genes, ras/physiology , fas Receptor/biosynthesis , fas Receptor/physiology , 3T3 Cells/metabolism , Animals , Cytokines/pharmacology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation/physiology , Humans , Mice , Mice, Inbred C3H , RNA, Messenger/metabolism , Sensitivity and Specificity , Signal Transduction/physiology , Transformation, Genetic/physiology , Up-Regulation/drug effects , fas Receptor/genetics
9.
J Immunol ; 160(11): 5465-74, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9605149

ABSTRACT

IL-12 is a potent immunoregulatory cytokine that has been shown to mediate tumor regression in a variety of tumor models. We describe the construction of AdCMV-IL-12, a recombinant adenovirus that encodes both subunits of IL-12 under transcriptional control of the CMV promoter. This recombinant virus efficiently infects a wide variety of cell types leading to the production of high levels of biologically active IL-12. Because the liver is a primary site of infection after i.v.-administered adenovirus, we tested the therapeutic efficacy of this virus in a murine hepatic metastasis tumor model. Systemic administration of AdCMV-IL-12 dramatically inhibited the formation of 3-day Renca hepatic metastases (mean of 16 metastases per liver) compared with the control virus AdCMV-betagal (mean of 209) or vehicle alone (mean of 272). Histologic analysis indicated that metastatic growth inhibition was accompanied by a dramatic perivascular infiltrate consisting of T cells, macrophages, and neutrophils. Therapeutic efficacy was not diminished in animals depleted of CD4+ or CD8+ T cells, or in SCID mice, even after NK cell ablation. In the latter case, a hepatic perivascular infiltrate composed of macrophages and neutrophils was observed after AdCMV-IL-12-treatment, while numerous activated Kupffer cells were noted in the hepatic parenchyma. Analysis of therapy-induced changes in hepatic gene expression demonstrated increased levels of IP-10 and Mig RNAs, but no increase in iNOS, Fas, or FasL RNA levels was observed. Our data suggest a model of metastatic growth inhibition mediated by nonlymphocyte effector cells including macrophages and neutrophils and that may involve anti-angiogenic chemokines.


Subject(s)
Adenoviridae/genetics , Interleukin-12/genetics , Killer Cells, Natural/immunology , Liver Neoplasms/secondary , Liver Neoplasms/therapy , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adenocarcinoma , Adenoviridae/immunology , Animals , Cell Movement/immunology , Gene Expression Regulation, Viral/immunology , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Injections, Intravenous , Interleukin-12/administration & dosage , Interleukin-12/biosynthesis , Kidney Neoplasms , Leukocytes, Mononuclear/pathology , Liver/pathology , Liver Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Tumor Cells, Cultured , Viral Vaccines/genetics
10.
J Immunother ; 21(2): 95-108, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9551360

ABSTRACT

Crosslinking of CD28 receptors on resting T lymphocytes by B7 costimulatory molecules expressed by antigen-presenting cells (APCs) plays a critical role in T-cell activation. Human melanomas express major histocompatibility complex (MHC)-restricted tumor-associated antigens that can be recognized by cytotoxic T lymphocytes (CTL), yet they remain poorly immunogenic. One mechanism for the failure of T-cell response is the lack of expression of costimulatory molecules by human melanoma cells. We have transfected the B7-1 gene into three HLA-A2-expressing human melanoma cell lines, and studied their capacity to stimulate primary human T cells. B7-expressing melanoma cells were excellent inducers of T-cell proliferation, cytokine production, and cytolytic activity in allogeneic mixed lymphocyte cultures through a process dependent on the function of the T-cell receptor as well as interactions between B7:CD28, CD2:LFA-3, and LFA-1:ICAM-1. Subset analysis demonstrated that CD4+ T cells or addition of exogenous interleukin-2 was required for the induction of CD8+ CTL. Untransfected parental melanoma cells were inert as APCs in these cultures. Rotating stimulation of T cells with the three B7-expressing cell lines led to the generation of T-cell lines that were cytolytic for HLA-A2+ melanoma cells and other HLA-A2+ targets that were pulsed with HLA-A2-restricted MART-1 peptides. These data demonstrate that expression of B7-1 by human melanoma cells converts them into effective APCs for the in vitro induction of MHC-restricted, melanoma-specific CTL.


Subject(s)
B7-1 Antigen/immunology , HLA-B7 Antigen/immunology , Lymphocyte Activation , Melanoma/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , B7-1 Antigen/genetics , Cell Communication , Cell Membrane , Cells, Cultured/immunology , Cytokines/metabolism , HLA-A2 Antigen/immunology , HLA-B7 Antigen/genetics , Humans , Lymphocyte Culture Test, Mixed , MART-1 Antigen , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transfection/immunology , Tumor Cells, Cultured/immunology
11.
Am J Clin Pathol ; 109(1): 90-5, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9426523

ABSTRACT

We describe the clinical and pathologic features of a lymphoepithelioma-like carcinoma (LELC) that originated in the hepatobiliary system. A woman, aged 71 years, was first seen with a noncholangiolar adenocarcinoma with lymphoid stroma, which was discovered by open liver biopsy in 1993. In 1995, retroperitoneal and peripancreatic lymph nodes were involved by LELC. There currently is no evidence of distant metastasis outside the hepatobiliary peripancreatic region. Review of the biopsy material revealed a well-differentiated adenocarcinoma with transition into LELC. Epstein-Barr virus (EBV) transcripts were expressed in all histologic phases of the tumor by in situ hybridization using immunoalkaline phosphatase-labeled oligonucleotide probes for EBV-encoded RNA 1 on formalin-fixed, paraffin-embedded sections. Polymerase chain reaction analysis for EBV nuclear antigen 2 was consistent with EBV strain type A. The LMP-1 gene was found to be wild type by polymerase chain reaction analysis. To our knowledge, this is the first report of a primary hepatobiliary adenocarcinoma associated with EBV infection that transformed into an undifferentiated LELC.


Subject(s)
Adenocarcinoma/virology , Herpesvirus 4, Human/isolation & purification , Liver Neoplasms/virology , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Aged , Antigens, Viral/genetics , Biomarkers/analysis , Biopsy , Female , Humans , Immunohistochemistry , In Situ Hybridization , Liver Neoplasms/chemistry , Liver Neoplasms/pathology , Lymph Nodes/chemistry , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphatic Metastasis , Polymerase Chain Reaction , RNA, Viral/analysis , Viral Matrix Proteins/genetics
12.
Article in English | MEDLINE | ID: mdl-18255936

ABSTRACT

A number of active prediction planning and execution (APPE) systems have recently been proposed for robotic interception of moving objects. The cornerstone of such systems is the selection of a robot-object rendezvous-point on the predicted object trajectory. Unlike tracking-based systems, which minimize the state difference between the object and the robot at each control period, in this methodology the robot is sent directly to the selected rendezvous-point. A fine-motion tracking strategy would then be employed for grasping the moving object. Herein, a novel strategy for selecting the optimal (earliest) rendezvous-point is presented. For objects with predictable trajectories, this is a significant improvement over previous APPE strategies which select the rendezvous-point from a limited number of non-optimally chosen candidates.

13.
FEMS Microbiol Lett ; 155(2): 217-22, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9351204

ABSTRACT

A cryptic plasmid from Xylella fastidiosa strain ATCC 35868 was cloned, sequenced, and the sequence entered into GenBank (U71220). The plasmid is 1296 nucleotides in length with 55% GC content and three open reading frames. A plasmid with sequence homology was found in only one other strain of X. fastidiosa, ATCC 35878. Searches of the GenBank reveal nucleotide sequence homology with plasmid pNKH43 from Stenotrophomonas maltophilia, and amino acid sequence homology with phage Pf3 from Pseudomonas aeruginosa, plasmid pAP12875 from Acetobacter pasteurianus, and plasmid pVT736-1 from Actinobacillus actinomycetemcomitans.


Subject(s)
Gram-Negative Bacteria/genetics , Plasmids , Amino Acid Sequence , Base Sequence , Escherichia coli/genetics , Molecular Sequence Data , Open Reading Frames , Sequence Homology, Amino Acid
14.
J Immunol ; 158(6): 2745-55, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058809

ABSTRACT

Signaling through the CD28 molecule on T cells by its natural ligand, B7, on APCs has recently been shown to require the presence of an active phosphatidylinositol 3-kinase pathway to mediate some of its costimulatory activities (1-7). Using the phosphatidylinositol 3-kinase inhibitor, wortmannin (WN) (8), on human and murine T cells, we have inhibited B7-1-mediated T cell activation and induced Ag-specific tolerance. The addition of WN and/or the B7-1 antagonist, CTLA4Ig, to primary human T cell cultures stimulated with B7-1-transfected allogeneic melanoma cell lines inhibited the generation of alloantigen-specific proliferative and cytolytic responses in vitro. Subsequent examination of these WN- and CTLA4Ig-treated primary T cell cultures revealed that these lymphocyte populations were tolerized to rechallenge with the priming alloantigens in secondary cultures in the absence of additional inhibitor(s). However, reactivity to a third party allogeneic stimulator remained intact. This WN-induced tolerance was reversed by the addition of high dose IL-2, but not IL-4 or IL-7, to the primary cultures, indicating that T cell anergy, not deletion, was responsible for this phenomenon. In vivo studies using a murine graft-vs-host disease (GVHD) model demonstrated that WN treatment of allogeneic donor lymphocytes in vitro failed to generate a significant GVHD in irradiated mouse recipients compared with control allogeneic donor lymphocytes. These findings suggest potentially novel therapeutic strategies for the prevention of GVHD.


Subject(s)
Androstadienes/pharmacology , Epitopes/immunology , Immune Tolerance/drug effects , Immunoconjugates , Isoantigens/immunology , Lymphocyte Activation/drug effects , T-Lymphocytes/immunology , Abatacept , Androstadienes/therapeutic use , Antigens, CD , Antigens, Differentiation/pharmacology , CTLA-4 Antigen , Clonal Anergy/drug effects , Dose-Response Relationship, Immunologic , Graft vs Host Disease/mortality , Graft vs Host Disease/prevention & control , Humans , Immunosuppressive Agents/pharmacology , Interleukin-2/pharmacology , Interleukin-4/pharmacology , Interleukin-7/pharmacology , Lymphocyte Culture Test, Mixed , T-Lymphocytes/drug effects , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Wortmannin
16.
Cancer Res ; 56(24): 5638-46, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8971169

ABSTRACT

One potential avenue for future cancer therapy involves the specific targeting of effector genes to cancer cells throughout the body, including distant metastatic sites. As a first step toward this goal, we tested the ability of the transcriptional regulatory elements of the human and mouse tyrosinase genes to promote high levels of pigment cell-specific transcription. A construct consisting of 209 bp of the human tyrosinase promoter linked to two enhancer elements was demonstrated to drive high-level, melanoma-specific expression of a beta-galactosidase (beta-gal) reporter gene in transient transfection assays. In studies of the murine tyrosinase promoter region, constructs containing up to 2500 bp of the 5' regulatory region were found to have very low transcriptional activity in murine melanoma cells. However, as with the human system, addition of two tandem repeats of an upstream enhancer element resulted in high levels of lineage-specific transcriptional activation. The murine tyrosinase promoter-enhancer expression cassette was introduced into the E1 region of a recombinant adenovirus to generate the virus AdmTyr-beta gal. This virus grows to high titer and maintains transcriptional specificity for pigment cell lineages. Strikingly, AdmTyr-beta gal is extremely active in human melanoma cells, in some cases exceeding the transcriptional activity of a cytomegalovirus promoter-driven recombinant beta-gal virus. Tissue specificity of gene expression is maintained, with very low levels observed in tumors and primary human cells derived from other lineages. These data provide evidence that it is possible to target human melanoma cells with great efficiency and specificity using high-titer recombinant adenovirus vectors.


Subject(s)
Genetic Vectors/genetics , Melanoma/enzymology , Monophenol Monooxygenase/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , beta-Galactosidase/metabolism , Adenoviridae/genetics , Animals , Enzyme Activation/genetics , Genes, Reporter , Humans , Melanoma/genetics , Mice , Monophenol Monooxygenase/metabolism , Tumor Cells, Cultured , beta-Galactosidase/genetics
17.
J Immunother Emphasis Tumor Immunol ; 19(5): 364-74, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8941876

ABSTRACT

We performed a prospective, randomized study to determine whether subcutaneous administration of interleukin-2 (IL-2) in combination with an autologous renal cell vaccine is feasible and can potentiate antitumor immunity. Seventeen patients with metastatic renal cell carcinoma underwent surgical resection with preparation of an autologous tumor cell vaccine. Patients were vaccinated intradermally twice at weakly intervals with 10(7) irradiated tumor cells plus bacillus Calmette-Guérin, and once with 10(7) tumor cells alone. Patients were randomized to one of three groups: no adjuvant IL-2, low-dose IL-2 (1.2 x 10(6) IU/m2), or high-dose IL-2 (1.2 x 10(7) IU/m2). IL-2 was administered subcutaneously on the day of vaccination and the subsequent 4 days. Immune response was monitored by delayed-type hypersensitivity (DTH) response to tumor cells as compared with normal autologous renal cells. Sixteen of 17 patients received vaccine therapy. Four patients developed cellular immunity specific for autologous tumor cells as measured by DTH responses; two had received no IL-2 and two had received high-dose IL-2. There were two partial responses (PR) noted, both in patients who received high-dose IL-2. One responding patient was DTH(+) and one was negative. A third patient who was DTH(+) after vaccination with no IL-2 had a dramatic PR after receiving IL-2 subcutaneously in a subsequent protocol. Prospective testing of response to recall antigens indicated that only 5 of 12 tested patients were positive, including both clinical responders. These data suggest that subcutaneously administered adjuvant IL-2 does not dramatically augment the immunologic response to autologous renal cell vaccines as determined by the development of tumor-specific DTH response.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Carcinoma, Renal Cell/immunology , Chemotherapy, Adjuvant , Humans , Hypersensitivity, Delayed/immunology , Immunotherapy, Adoptive , Injections, Subcutaneous , Kidney Neoplasms/immunology
18.
J Clin Oncol ; 14(8): 2234-41, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8708712

ABSTRACT

PURPOSE: Although high-dose interleukin-2 (IL-2) can produce durable remissions in a subset of responding patients with renal cell carcinoma (RCC), this occurs in the setting of significant toxicity. The purpose of this study is to define the maximum-tolerated dosage (MTD) of IL-2 and interferon alfa-2a (IFN alpha-2a) that can be administered chronically on an outpatient basis. PATIENTS AND METHODS: Fifty-three patients with advanced cancer of variable histology with good prognostic features were treated in six cohorts. Patients in cohorts one through five received IL-2 (1.5 or 3.0 x 10(6) million units (mU)/m2) Monday through Friday and IFN alpha-2a (1.5 or 3 x 10(6) mU/m2) daily for a 4-week cycle. In cohort six, IFN alpha-2a was given three times a week. Immunologic monitoring, including serum levels of soluble IL-2 receptor (sIL-2R) and neopterin, flow cytometry, and natural killer cell (NK) activity, were measured. Patients were evaluated for toxicity, response, and survival. RESULTS: Almost all patients developed grade I/II toxicities commonly associated with cytokine therapy. Symptoms were most severe with the first treatment of each week. Dose-limiting toxicities included grade III fatigue, hypotension, and creatinine elevations. The MTD was 1.5 mU/m2 daily x 5 given subcutaneously repeated weekly for IL-2 and 1.5 mU/m2 daily subcutaneously (dose level 3) for IFN. Six of 25 assessable patients with RCC (24%) achieved a partial response (PR), including four of eight patients who were previously untreated. There were no objective responses in patients with other tumors, including 12 melanoma patients. CONCLUSION: IL-2 and IFN alpha-2a can be given with tolerable toxicities on an outpatient basis and shows significant activity in patients with metastatic RCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Interferon-alpha/therapeutic use , Interleukin-2/therapeutic use , Neoplasms/therapy , Adult , Aged , Antineoplastic Agents/adverse effects , Biopterins/analogs & derivatives , Biopterins/blood , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Cohort Studies , Female , Humans , Injections, Subcutaneous , Interferon alpha-2 , Interferon-alpha/adverse effects , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Killer Cells, Natural/immunology , Male , Middle Aged , Neoplasms/immunology , Neopterin , Receptors, Interleukin-2/metabolism , Recombinant Proteins , Remission Induction
19.
J Immunother Emphasis Tumor Immunol ; 19(4): 296-308, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8877723

ABSTRACT

The adoptive transfer of anti-CD3-stimulated T killer (T-AK) cells was tested with different bolus and infusional interleukin-2 (IL-2) regimens, and anti-CD3 stimulation procedures to determine immunologic and antitumor effects in patients with a variety of advanced cancers. Indium-111 labeling was used to observe traffic patterns of the infused T-AK. Autologous peripheral blood mononuclear cells were obtained by leukapheresis. Cyclophosphamide (300 mg/m2) was given to most patients immediately after leukapheresis. The harvested cells were activated ex vivo with anti-CD3 overnight or for 4 days, at which time cells were reinfused and an IL-2 regimen was begun. Treatment was repeated 28 days later. This treatment regimen induced significant increases in leukocytes, lymphocytes, and eosinophils in patients in most treatment cohorts. Circulating lymphocytes were predominantly CD3+ T cells with preferential expansion of the CD8+ subset. Patients receiving cells stimulated in vitro for 4 days had significant T-cell lymphocytosis with either infusional or bolus plus infusional IL-2 regimens. T-cell viability was decreased in culture after a second 4-day stimulation with anti-CD3 at day 28; this decrease could be prevented by adding IL-2 to the culture media. Cells stimulated overnight required both bolus and infusional IL-2 to show an atypical lymphocytosis in vivo. Overnight-stimulated T-AK did not show decreases in in vitro viability at the day 28 restimulation. Indium-III-labeled cells trafficked to the liver, spleen, and bone marrow. No increase in uptake was observed in tumor deposits. There were 2 patients with partial responses, 5 with minor responses, 19 with stable disease, and 88 with progressive disease. The length of in vitro anti-CD3 stimulation, and the dose and timing of IL-2 administration in vivo results in different circulating leukocyte populations after adoptive T-AK infusion. Generally, the CD8+ T-cell subset was preferentially expanded by this treatment approach. Repeated ex vivo stimulation with anti-CD3 may cause cell death.


Subject(s)
Antibodies, Monoclonal/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive , Interleukin-2/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocyte Subsets/drug effects , Adolescent , Adult , Aged , Cell Movement/drug effects , Cell Movement/immunology , Dose-Response Relationship, Immunologic , Drug Administration Routes , Drug Administration Schedule , Female , Humans , Interleukin-2/adverse effects , Lymphocyte Activation/drug effects , Male , Middle Aged , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/transplantation
20.
Blood ; 87(1): 202-10, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8547643

ABSTRACT

Stimulation of B lymphocytes through the Ig receptor initiates a cascade of biochemical changes, which can ultimately lead to either activation and growth, or cell-cycle arrest and cell death. One of the critical events that occurs in both cases is the activation of tyrosine kinases, and the resulting phosphorylation of a variety of proteins on tyrosine residues. In this report we identify one of the substrates of phosphorylation as the 85-kD subunit of the enzyme phosphatidylinositol-3 kinase (PI3K), and show that both anti-IgM and anti-IgD stimulation results in an increase in the anti-phosphotyrosine-precipitable PI3K activity. Furthermore, we show that the potent and specific inhibitor of PI3K, Wortmannin, can completely abrogate anti-Ig-mediated growth inhibition without affecting tyrosine kinase induction or protein kinase C (PKC) activation. Treatment of intact cells with Wortmannin results in an irreversible decrease in anti-Ig-induced PI3K activity, suggesting that the effect of Wortmannin on anti-Ig-mediated growth inhibition is caused by its inactivation of PI3K activity. Taken together, these data show that activation of PI3K is a critical component of the anti-Ig-initiated signaling cascade that leads to growth inhibition of human B lymphoma cells.


Subject(s)
B-Lymphocytes/pathology , Immunoglobulin D/physiology , Immunoglobulin M/physiology , Lymphoma, Large B-Cell, Diffuse/pathology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Protein Processing, Post-Translational , Receptors, Antigen, B-Cell/physiology , Signal Transduction/physiology , Androstadienes/pharmacology , Antibodies, Anti-Idiotypic/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Cell Division/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Ligands , Neoplasm Proteins/physiology , Phosphatidylinositol 3-Kinases , Phosphorylation , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Antigen, B-Cell/antagonists & inhibitors , Tumor Cells, Cultured , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...