Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
2.
Hernia ; 20(1): 161-70, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26545361

ABSTRACT

BACKGROUND: Decellularized porcine small intestinal submucosa (SIS) is a biological scaffold used surgically for tissue repair. Here, we demonstrate a model of SIS as a scaffold for human adipose-derived stem cells (ASCs) in vitro and apply it in vivo in a rat ventral hernia repair model. STUDY DESIGN: ASCs adherence was examined by confocal microscopy and proliferation rate was measured by growth curves. Multipotency of ASCs seeded onto SIS was tested using adipogenic, chondrogenic, and osteogenic induction media. For in vivo testing, midline abdominal musculofascial and peritoneal defects were created in Sprague-Dawley rats. Samples were evaluated for tensile strength, histopathology and immunohistochemistry. RESULTS: All test groups showed cell adherence and proliferation on SIS. Fibronectin-treated scaffolds retained more cells than those treated with vehicle alone (p < 0.05). Fresh stromal vascular fraction (SVF) pellets containing ASCs were injected onto the SIS scaffold and showed similar results to cultured ASCs. Maintenance of multipotency on SIS was confirmed by lineage-specific markers and dyes. Histopathology revealed neovascularization and cell influx to ASC-seeded SIS samples following animal implantation. ASC-seeded SIS appeared to offer a stronger repair than plain SIS, but these results were not statistically significant. Immunohistochemistry showed continued presence of cells of human origin in ASC-seeded repairs at 1 month postoperation. CONCLUSION: Pretreatment of the scaffold with fibronectin offers a method to increase cell adhesion and delivery. ASCs maintain their immunophenotype and ability to differentiate while on SIS. Seeding freshly isolated SVF onto the scaffold demonstrated that minimally manipulated cells may be useful for perioperative surgical applications within the OR suite. We have shown that this model for a "living mesh" can be successfully used in abdominal wall reconstruction.


Subject(s)
Hernia, Ventral/surgery , Intestinal Mucosa/physiology , Intestine, Small/physiology , Mesenchymal Stem Cells/physiology , Tissue Scaffolds , Wound Healing/physiology , Animals , Cell Proliferation , Female , Hernia, Ventral/physiopathology , Herniorrhaphy/methods , Humans , Microscopy, Confocal , Models, Animal , Rats , Rats, Sprague-Dawley , Swine , Tensile Strength
3.
Ann Oncol ; 25(3): 592-598, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24401928

ABSTRACT

BACKGROUND: Nonpegylated liposomal doxorubicin liposomal doxorubicin, (Myocet™; Sopherion Therapeutics, Inc Canada, and Cephalon, Europe) (NPLD; Myocet(®)) in combination with trastuzumabHerceptin(®) (Hoffmann-La Roche) has shown promising activity and cardiac safety. We conducted a randomized phase III trial of first-line NPLD plus trastuzumab and paclitaxel (Pharmachemie B.V.) (MTP) versus trastuzumab plus paclitaxel (TP) in patients with human epidermal growth factor 2 receptor (HER2)-positive metastatic breast cancer. PATIENTS AND METHODS: Patients were randomly assigned to NPLD (M, 50 mg/m(2) every 3 weeks for six cycles), trastuzumab (T, 4 mg/kg loading dose followed by 2 mg/kg weekly), and paclitaxel (P, 80 mg/m(2) weekly) or T + P at the same doses until progression or toxicity. The primary efficacy outcome was progression-free survival (PFS). RESULTS: One hundred and eighty-one patients were allocated to receive MTP, and 183 to TP. Median PFS was 16.1 and 14.5 months with MTP and TP, respectively [hazard ratio (HR) 0.84; two-sided P = 0.174]. In patients with estrogen receptor (ER)- and progesterone receptor (PR)-negative tumors, PFS was 20.7 and 14.0 months, respectively [HR 0.68; 95% confidence interval (CI) 0.47-0.99]. Median overall survival (OS) was 33.6 and 28.9 months with MTP and TP, respectively (HR 0.79; two-sided P = 0.083). In ER- and PR-negative tumors, OS was 38.2 and 27.9 months, respectively (HR 0.63; 95% CI 0.42-0.93). The frequency of adverse events was higher with MTP, but there was no significant difference in cardiac toxicity between treatment arms. CONCLUSION(S): The trial failed to demonstrate a significant clinical improvement with the addition of M to TP regimen. The clinical benefit observed in an exploratory analysis in the ER- and PR-negative population deserves consideration for further clinical trials. CLINICAL TRIAL NUMBER: NCT00294996.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Paclitaxel/therapeutic use , Receptor, ErbB-2/metabolism , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Drug Administration Schedule , Female , Humans , Neoplasm Metastasis/drug therapy , Paclitaxel/adverse effects , Polyethylene Glycols/adverse effects , Polyethylene Glycols/therapeutic use , Prospective Studies , Trastuzumab , Treatment Outcome
4.
Scand J Immunol ; 66(4): 402-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17850584

ABSTRACT

Tumour infiltration by activated natural killer (A-NK) cells is a pre-requisite for tumour eradication by adoptive NK cell transfer. Extravasated A-NK cells do not always succeed in reaching the crucial target cell conjugation. Therefore, we wished to study A-NK cell locomotion and interactions with melanoma cells in a matrix environment (Matrigel) by electron, confocal and fluorescence microscopy. Two distinct patterns of A-NK cell-mediated matrix disintegration were revealed during incubation of tumour cells and A-NK cells in Matrigel: (1) A-NK cells pre-cultured for 5 days altered the homogeneous texture of the Matrigel, an initial microporous appearance became a loose filamentous meshwork by 24 h. Matrix degrading protease inhibitors could not fully prevent this, but could delay the process; and (2) A-NK cells pre-cultured for 6 days or more, instead formed large excavations in the Matrigel leaving the remaining matrix less affected compared to the effects by the younger A-NK cells. By histochemical staining with Cupromeronic Blue, the excavations were shown to contain proteoglycan material. Protease inhibitors had no discernable effect on the development of the excavations. The conspicuous capacity of A-NK cells to disintegrate extracellular matrix and the formation of large excavations seems only partially to depend on matrix-degrading proteases. Formation of extracellular proteoglycan material is suggested to facilitate A-NK cell locomotion within a matrix environment.


Subject(s)
Cell Movement/immunology , Collagen , Interleukin-2/immunology , Killer Cells, Lymphokine-Activated/immunology , Laminin , Melanoma, Experimental/immunology , Proteoglycans , Animals , Cell Line, Tumor , Coculture Techniques , Drug Combinations , Histocytochemistry , Humans , Indoles/chemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Organometallic Compounds/chemistry
5.
Histopathology ; 44(1): 54-63, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14717670

ABSTRACT

AIMS: Some experimental evidence suggests that in lung cancer, development, progression and an increased proliferation rate can be linked to apoptosis-related factors. In this study we evaluated the possible role of p53 and Bcl-2 gene family members as prognostic factors for non-small-cell lung cancer. METHODS AND RESULTS: We investigated the immunohistochemical expression of p53 and Bcl-2 gene family members (bax, Bcl-2 and Bcl-xL) in 94 non-small-cell lung cancer specimens to establish the role of these genes in lung cancer pathogenesis, and to evaluate their prognostic importance. The expression of Bcl-2 was correlated with a shorter patient survival time and with the nodal status of the neoplasm. We also found frequent over-expression of bax and Bcl-xL to be of no prognostic significance. Finally, we found no correlation between frequent detection of aberrant p53 protein and expression of either Bcl-2, bax or Bcl-xL or with patient survival time. CONCLUSIONS: This study confirms a relevant role for apoptosis-regulatory proteins in the pathogenesis of lung cancer, and highlights the possible role of Bcl-2 as a prognostic factor for this tumour.


Subject(s)
Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Female , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins/metabolism , Survival Rate , bcl-2-Associated X Protein , bcl-X Protein
6.
Anticancer Res ; 21(3B): 1697-704, 2001.
Article in English | MEDLINE | ID: mdl-11497249

ABSTRACT

BACKGROUND: The urokinase plasminogen activator (uPA) system has been implicated in cellular invasiveness of tumor cells and immune cells. Herein we provide evidence for the production by natural killer (NK) cells of both uPA and its receptor (uPAR). MATERIALS AND METHODS: Western blot analysis, RTPCR, casein/plasminogen zymography, and fluorescence microscopy were employed to detect uPA and uPAR on NK cells. NK cell invasiveness was examined using Matrigel invasion assays. RESULTS: NK cell uPA appeared at its characteristic molecular weights, is enzymatically active in casein/plasminogen zymography, and is recognized by monoclonal antibodies. uPAR was detected by RTPCR and fluorescence microscopy. Matrigel invasion assays demonstrated an active role of uPA in NK cell invasion. CONCLUSION: The uPA system contributes to extracellular matrix (ECM) degradation by NK cells, which may be essential for NK cell accumulation into metastases, and may be prerequisite for their killing of tumor cells following NK cell adoptive transfer.


Subject(s)
Extracellular Matrix/metabolism , Killer Cells, Natural/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , 3T3 Cells , Animals , Antibodies, Monoclonal/metabolism , Aprotinin/metabolism , Blotting, Western , Caseins/metabolism , Cell Membrane/metabolism , Cell Movement , Collagen/metabolism , Drug Combinations , Humans , Laminin/metabolism , Mice , Microscopy, Fluorescence , Neoplasm Invasiveness , Phenotype , Phosphatidylinositol Diacylglycerol-Lyase , Plasminogen/metabolism , Proteoglycans/metabolism , RNA, Messenger/metabolism , Receptors, Urokinase Plasminogen Activator , Reverse Transcriptase Polymerase Chain Reaction , Type C Phospholipases/pharmacology , U937 Cells
7.
Anticancer Res ; 21(1A): 45-50, 2001.
Article in English | MEDLINE | ID: mdl-11299777

ABSTRACT

BACKGROUND: Previous studies have shown that MMP-8, the neutrophil collagenase, was expressed in neutrophils, chondrocytes and rheumatoid synovial fibroblasts. MATERIALS AND METHODS: We used semi-quantitative RT-PCR analysis, Western blotting, and immunofluorescence assays to determine the expression of MMP-8 in Jurkat T cells. RESULTS: We have determined the expression of MMP-8 from Jurkat cells and the down-regulation of its expression by genistein, a principal soy isoflavone. Genistein inhibited the invasion of Jurkat cells through a model basement membrane by about 75%, similar to the inhibition by BB-94, a synthetic MMP inhibitor. Genistein also down-regulated the expression of MMP-13, but slightly up-regulated the expression of TIMP-1 and TIMP-2. CONCLUSIONS: Our findings documented for the first time the expression of the neutrophil collagenase by a T-cell line. We also determined the inhibition of Jurkat cell invasion by genistein, which was in part mediated through the regulation of the expression of MMPs and TIMPs.


Subject(s)
Leukemia, T-Cell/enzymology , Leukemia, T-Cell/genetics , Matrix Metalloproteinase 8/physiology , Antineoplastic Agents/pharmacology , Collagenases/biosynthesis , Collagenases/genetics , Down-Regulation , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/physiology , Gene Expression Regulation, Neoplastic , Genistein/pharmacology , Humans , Jurkat Cells , Leukemia, T-Cell/pathology , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 8/biosynthesis , Matrix Metalloproteinase 8/genetics , Neoplasm Invasiveness , Tissue Inhibitor of Metalloproteinases/biosynthesis , Tissue Inhibitor of Metalloproteinases/genetics
8.
Histochem Cell Biol ; 115(1): 67-72, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11219610

ABSTRACT

In this article we report about the role that tumor structure and extracellular matrix (ECM) may play in immunotherapy and in gene therapy using adenoviruses. We performed studies in a rat model for colorectal cancer, CC531, and in specimens of human colorectal cancer. The tumors were composed of two compartments, tumor cell nests surrounded by stromal cells. ECM proteins were expressed in the stromal part, where the blood vessels were also located. Furthermore, in several tumors, the tumor cell nests were surrounded by basal membrane-like structures. Therefore, in vascular approaches to treat cancer, therapeutic agents on their route to tumor cells may be hampered by ECM to reach tumor cells. We found that immune cells were abundantly present in tumors from colorectal origin. These cells were, however, not found in direct contact with tumor cells, but mainly in the stromal part of the tumor. Adenoviruses, when intravascularly injected, did not reach tumor cells in the CC531 rat model. Tumor cells were only infected, and even then in limited numbers, in cases of intratumoral injection. We hypothesize that ECM in a tumor is a barrier both for immune cells and for adenoviruses to make direct contact with these tumor cells, and thus limits colorectal tumor therapy.


Subject(s)
Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Extracellular Matrix/metabolism , Genetic Therapy/methods , Adenoviridae/genetics , Aged , Aged, 80 and over , Animals , Antibodies, Neoplasm/immunology , Antibody Formation/genetics , Colorectal Neoplasms/therapy , Female , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Humans , Immunohistochemistry , Killer Cells, Natural , Lac Operon , Laminin/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Middle Aged , Rats , Rats, Wistar , T-Lymphocytes , Tissue Distribution
9.
In Vivo ; 14(5): 557-64, 2000.
Article in English | MEDLINE | ID: mdl-11125539

ABSTRACT

In this study, we describe rat NK cell-derived MMPs including membrane-type MMPs (MT-MMPs) and tissue inhibitors of MMP (TIMPs). RT-PCR analysis from cDNA of rat A-NK cells revealed mRNA for MMP-2, MMP-9, MMP-7, MMP-10, MMP-11, MMP-13, MT1-MMP, MT2-MMP, TIMP-1, and TIMP-2. The RNK-16 cells expressed mRNA for MMP-7, MMP-10, MMP-11, MT1-MMP, MT2-MMP, TIMP-1, and TIMP-2, in addition to MMP-3 and MMP-13. Western blot analysis confirmed proteins for MT1-MMP and MT2-MMP in RNK-16 cells. TIMP-1 in rat A-NK cells was present at molecular mass of 34-kDa protein which may represent a highly glycosylated form. Genistein, a natural isoflavone found in soybeans, inhibited proliferation of RNK-16 cells in dosage dependent manner. In addition, it down-regulated the expression of MMP-13, MT1-MMP, TIMP-1 and TIMP-2. Moreover, genistein greatly impaired the ability of RNK-16 cells to invade through a model basement membrane. This effect might be mediated by the observed down-regulation of MMP-13 and MT1-MMP.


Subject(s)
Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Killer Cells, Natural/enzymology , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Animals , Blotting, Western , Cell Division/drug effects , DNA Primers/chemistry , DNA, Complementary/analysis , Dose-Response Relationship, Drug , Down-Regulation , Killer Cells, Natural/drug effects , Male , Matrix Metalloproteinases/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinases/genetics , Tumor Cells, Cultured
10.
In Vivo ; 14(5): 565-70, 2000.
Article in English | MEDLINE | ID: mdl-11125540

ABSTRACT

We have previously investigated the role of the urokinase plasminogen activator (uPA) system in NK cell invasion. We have also studied NK cell-derived matrix metalloproteinases (MMPs) in extracellular matrix (ECM) degradation. We now report that both enzyme systems cooperate in NK cell invasion. Zymographic analyses detected uPA in RNK-16 cell conditioned media (CM) with the same molecular weights as the uPA we have previously shown to be associated with the rat NK cell urokinase plasminogen activator receptor. The combination of aprotinin, an inhibitor of plasmin, and Batimastat (BB94), an inhibitor of MMPs, in Matrigel invasion assays showed a more potent inhibitory effect on NK cell invasion than either inhibitor alone. Finally, a down regulation of uPA mRNA was noted following RNK-16 stimulation with collagen IV, fibronectin, and laminin.


Subject(s)
Killer Cells, Natural/enzymology , Lymphocytes, Tumor-Infiltrating/metabolism , Matrix Metalloproteinases/metabolism , Phenylalanine/analogs & derivatives , Urokinase-Type Plasminogen Activator/metabolism , Animals , Aprotinin/pharmacology , Collagen/pharmacology , Culture Media, Conditioned/chemistry , Down-Regulation , Fibronectins/pharmacology , Humans , Killer Cells, Natural/drug effects , Laminin/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Phenylalanine/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thiophenes/pharmacology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics
11.
In Vivo ; 14(5): 651-8, 2000.
Article in English | MEDLINE | ID: mdl-11125549

ABSTRACT

The circulatory pattern of IL-2 activated natural killer (A-NK) cells was studied in C57BL/6 mice bearing 10 day-old pulmonary and subcutaneous (s.c.) metastases of the B16 melanoma in order to evaluate the roles of the concentration of A-NK cells in the blood and of tumor blood flow on accumulation of A-NK cells in tumors. Kinetic studies of the presence of A-NK cells in peripheral blood after adoptive transfer revealed that these cells rapidly disappear from the blood. Via intravital microscopy of animals with exposed lung tissue, we have shown that the vast majority of transferred A-NK cells become efficiently arrested within the lung microcirculation at their first encounter with this organ, thereby explaining the fast disappearance of the cells from the bloodstream. Despite the low number of A-NK cells circulating in the blood, systemically injected A-NK cells (20 million per mouse) localized significantly (70-80 million cells/g) into most pulmonary metastases within 8-16 hours. In contrast, very few A-NK cells (< 0.2 million cells/g) were found in the s.c. metastases. Based on measurements of tumor blood flow (showing a classic inverse relationship between tumor size and tumor blood flow) and the blood concentration of A-NK cells, we estimated the highest intratumoral density of A-NK cells that theoretically can be generated by A-NK cells transported to the tumor by way of the blood. In s.c. tumors, the observed density of A-NK cells was at all times lower (10-50 fold) than the estimated density, indicating that only a few percent of the A-NK cells arriving at these tumors become retained in them. In contrast, the observed density of A-NK cells in pulmonary metastases was at all times higher (2-3 fold) than the estimated density. This finding indicates that A-NK cells might not reach the pulmonary metastases solely by way of the blood stream. In conclusion, i.v. injected A-NK cells become immediately entrapped in the lungs and, consequently, circulate poorly. While lung metastases become significantly infiltrated by i.v. injected A-NK cells, metastases in organs down-stream from the lungs become poorly infiltrated. We hypothesize that only a part of the A-NK cells found in lung metastases 8-16 hours following injection reach these metastases by way of the blood-vascular system. They might also migrate into the metastases from the surrounding normal lung tissue.


Subject(s)
Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Lung Neoplasms/blood supply , Melanoma, Experimental/blood supply , Skin Neoplasms/blood supply , Animals , Cell Count , Immunotherapy, Adoptive , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/pathology , Killer Cells, Lymphokine-Activated/transplantation , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Microcirculation , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Spleen/cytology , Spleen/drug effects
12.
In Vivo ; 14(5): 625-9, 2000.
Article in English | MEDLINE | ID: mdl-11125547

ABSTRACT

IFN-gamma is a cytokine that regulates various functions of the immune system. The major producers of IFN-gamma are T cells and NK cells. 2B4 is a novel activating receptor expressed on all human NK cells, a subset of CD8+ T cells, monocytes and basophils. Activation of human NK cells through surface 2B4 enhances NK cell cytolytic function and secretion of IFN-gamma. We have examined the regulation of IFN-gamma production by the human NK cell line YT upon activation through surface 2B4. Our data indicate that ligation of surface 2B4 by mAb C1.7, that specifically recognizes 2B4, induces transcriptional activation of IFN-gamma. Partial inhibition of transcription did not prevent the transcriptional upregulation of IFN-gamma. S1 nuclease protection analysis indicated that transcriptional activation as well as mRNA stability may account for the increased production of IFN-gamma by human NK cells following 2B4 stimulation.


Subject(s)
Antigens, CD , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic , Signal Transduction , Actins/genetics , Actins/metabolism , Cell Line , DNA Primers/chemistry , Dactinomycin/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation , RNA, Messenger/metabolism , Signaling Lymphocytic Activation Molecule Family , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Up-Regulation
14.
Immunology ; 100(3): 378-83, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10929061

ABSTRACT

2B4 is a surface molecule found on all human natural killer (NK) cells, a subset of CD8+ T cells, monocytes and basophils. It was originally identified on mouse NK cells and the subset of T cells that mediate non-major histocompatibility complex (MHC)-restricted killing. Recently,9 we have cloned the human homologue of 2B4 (h2B4) and found h2B4 to also mediate non-MHC-restricted cytotoxicity. In this study, we examine h2B4 in regulating various functions of NK cells using a human NK cell line YT, with monoclonal antibody (mAb) C1.7, an antibody that specifically recognizes h2B4. Ligation of surface 2B4 with mAb C1.7 increases YT's ability to destroy tumour cells. In the presence of mAb C1.7, the production of interferon-gamma (IFN-gamma) by YT cells is greatly enhanced. Engagement of surface 2B4 by mAb C1.7 downregulates the expression of h2B4 at the cell surface as well as the expression of h2B4 mRNA. Also, signalling through h2B4 causes the increased expression of matrix metalloproteinase-2, a member of the matrix degrading proteinase family. Thus, in addition to modulating cytolytic function and cytokine production of NK cells, activation through surface 2B4 may play a role in upregulating the machinery for degradation of extracellular matrices to promote invasion of the tumour by NK cells.


Subject(s)
Antigens, CD , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Membrane Glycoproteins/immunology , Receptors, Immunologic , Animals , Antibodies, Monoclonal/immunology , Cell Line , Gene Expression Regulation/immunology , Humans , Interferon-gamma/biosynthesis , Matrix Metalloproteinase 2/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , RNA, Messenger/genetics , Signaling Lymphocytic Activation Molecule Family , Tumor Cells, Cultured , Up-Regulation/immunology
15.
FASEB J ; 14(10): 1400-10, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10877833

ABSTRACT

Urokinase plasminogen activator (uPA) plays an important role in the progression of several malignancies including breast cancer. We have identified a noncompetitive antagonist of the uPA-uPAR interaction derived from a nonreceptor binding region of uPA (amino acids 136-143). This 8-mer capped peptide (A6) inhibited breast cancer cell invasion and endothelial cell migration in a dose-dependent manner in vitro without altering cell doubling time. Intraperitoneal administration of A6 resulted in a significant inhibition of tumor growth and suppressed the development of lymph node metastases in several models of breast cancer cell growth and metastasis. Large areas of tumor necrosis and extensive positive staining by TUNEL were observed on histological and immunohistochemical analysis of experimental tumor sections from A6-treated animals. A6 treatment also resulted in a decrease in factor VIII-positive tumor microvessel hot-spots. These results identify a new epitope in uPA that is involved in the uPA-uPAR interaction and indicate that an antagonist based on this epitope is able to inhibit tumor progression by modulating the tumor microenvironment in the absence of direct cytotoxic effects in vivo.


Subject(s)
Breast Neoplasms/drug therapy , Peptide Fragments/pharmacology , Urokinase-Type Plasminogen Activator/pharmacology , Amino Acid Sequence , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Movement/drug effects , Female , Humans , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Necrosis , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/prevention & control , Peptide Fragments/chemistry , Rats , Rats, Inbred F344 , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism
16.
Cancer Epidemiol Biomarkers Prev ; 9(6): 609-17, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10868697

ABSTRACT

After an initial series of experiments indicated that early responses of B lymphocytes were important in controlling tumor metastases in two rat models of cancer (N. Quan et al., Cancer Res., 59: 1080-1089, 1999), the present study assessed whether differences in the number of B lymphocytes that are normally present in different individual rats before any tumor development could predict tumor growth, metastases, and length of survival when tumor challenge subsequently occurred. Repeated baseline measures of several circulating lymphocyte subtypes (i.e., natural killer, B, CD4+, CD8+ lymphocytes) were made in individual inbred WAG rats before any introduction of tumor cells, and stable baselines for these subtypes were found. Animals were then injected with 2 x 10(6) CC531 tumor cells (a syngeneic tumor) into the leg, and the size of the resulting primary tumor measured. Primary tumors were surgically removed 6-7 weeks after tumor-cell injection, and animals then followed until death from metastases. In two experiments, the size of the primary tumor as well as the length of time that animals survived correlated with the pretumor percentage of certain lymphocyte subtypes in peripheral blood before tumor-cell injection. Baseline percentage of B lymphocytes was significantly negatively correlated with the size of the primary tumor and was positively correlated with the duration of survival. Baseline percentage of CD4+ lymphocytes showed the opposite relationship, being positively correlated with tumor size and negatively correlated with survival time, although these correlations were lower than those for B lymphocytes. Percent B lymphocytes in circulation also declined during tumor development. In summary, a high percentage of endogenous peripheral blood B lymphocytes predicted growth of smaller primary tumors and longer survival after experimental tumor induction in a rat model, further suggesting that B lymphocytes are involved in protection against development of certain tumors.


Subject(s)
Adenocarcinoma/immunology , B-Lymphocytes , CD4-Positive T-Lymphocytes , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Animals , CD8-Positive T-Lymphocytes , Disease Models, Animal , Female , Killer Cells, Natural , Lymphocyte Count , Lymphocyte Subsets , Neoplasm Metastasis/immunology , Prognosis , Rats , Rats, Inbred Strains , Survival Analysis , Tumor Cells, Cultured
17.
J Immunol ; 164(11): 5883-9, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10820269

ABSTRACT

We have previously documented that rat IL-2-activated NK (A-NK) cells produce matrix metalloproteinase-2 (MMP-2) and MMP-9. In this study, we describe mouse A-NK cell-derived MMPs, including MT-MMPs, and also TIMPs. RT-PCR analysis from cDNA of mouse A-NK cells revealed mRNA for MMP-2, MMP-9, MMP-11, MMP-13, MT1-MMP, MT2-MMP, TIMP-1, and TIMP-2. MMP-2 and MMP-9 expression was confirmed by gelatin zymography. Moreover, we report for the first time that MT-MMPs are expressed by NK cells, i.e., large granular lymphocytes as determined by both RT-PCR and Western blots. TIMP-1 expression was detected as a 29-kDa protein in Western blots. It is intriguing that TIMP-2 protein from A-NK cells was also detected as a 29-kDa protein, which is clearly different from the previously reported molecular mass of 21 kDa in mouse and human cells. In addition, inhibition of MMPs by BB-94, a selective inhibitor of MMP, significantly inhibited the ability of mouse A-NK cells to migrate through Matrigel, a model basement membrane. Taken together, these findings suggest that A-NK cells may therefore use multiple MMPs in various cellular functions, including degradation of various extracellular matrix molecules as they extravasate from blood vessels and accumulate within cancer metastases following their adoptive transfer.


Subject(s)
Interleukin-2/physiology , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Lymphocyte Activation , Matrix Metalloproteinases/isolation & purification , Matrix Metalloproteinases/metabolism , Membrane Proteins/isolation & purification , Tissue Inhibitor of Metalloproteinases/isolation & purification , Animals , Blotting, Western , Cell Membrane/enzymology , Cell Membrane/immunology , Cell Movement/immunology , Cell-Free System/enzymology , Cell-Free System/immunology , Cells, Cultured , DNA, Complementary/analysis , Diffusion Chambers, Culture , Electrophoresis, Polyacrylamide Gel , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinases/physiology
18.
Arch Pathol Lab Med ; 124(5): 766-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10782166

ABSTRACT

We report the first 2 cases, to our knowledge, of retroperitoneal cysts with features of mesothelial differentiation that clinically mimic renal masses. The first lesion occurred in a 71-year-old man who presented with flank pain. Ultrasound and magnetic resonance imaging studies showed a unilocular cystic structure arising from the upper pole of the left kidney. The second lesion was in a 44-year-old woman who presented with left flank pain. Imaging studies revealed an 8-cm hemorrhagic cyst at the lower pole of the left kidney. Histologic examination of the nephrectomy specimens in each case revealed a unilocular cyst with intracystic and pericystic hemorrhage. In each case, the cyst was lined by a single layer of cells with ample eosinophilic cytoplasm and benign nuclear features without mucinous or müllerian differentiation. Histochemical staining showed Alcian blue positivity on the cell surface, which was sensitive to hyaluronidase digestion. Intracytoplasmic mucin, however, was not detected. Immunostaining showed that the cyst lining cells were positive for keratin, vimentin, HBME-1, WT1, and thrombomodulin but negative for carcinoembryonic antigen, B72.3, Leu-M1, and BerEP4. The first case was positive for calretinin, whereas the second was negative. These findings support the mesothelial nature of the cysts.


Subject(s)
Cysts/pathology , Neoplasms, Mesothelial/pathology , Retroperitoneal Neoplasms/pathology , Adult , Aged , Antigens, Surface/metabolism , Cysts/metabolism , Cysts/surgery , Diagnosis, Differential , Female , Hemorrhage/etiology , Humans , Immunohistochemistry , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Male , Neoplasms, Mesothelial/metabolism , Neoplasms, Mesothelial/surgery , Nephrectomy , Pain/etiology , Retroperitoneal Neoplasms/metabolism , Retroperitoneal Neoplasms/surgery
19.
In Vivo ; 14(1): 101-4, 2000.
Article in English | MEDLINE | ID: mdl-10757065

ABSTRACT

Adoptively transferred IL-2 activated NK (A-NK) cells selectively accumulate within tumor metastases which recommends them as vehicles for locoregional drug delivery. Zyn-Linkers are membrane-binding lipophilic dyes which can be coupled by a variety of conjugation chemistries to therapeutic agents. We have previously demonstrated that A-NK cells labeled with PKH26 are able to accumulate within established B16 melanoma pulmonary metastases by 16 h at a concentration of over 600 cells/mm2 of tumor tissue (Basse et al. J. Exp. Med. 174: 479 1991). Zyn-205 is a prodrug in which doxorubicin is attached to a similar Zyn-Linker through an acid-sensitive bond. We have optimized the ex vivo labeling conditions and found that a 10 min incubation with 25 microM Zyn-205 results in the uptake of over 10(8) drug molecules per cell with no effect on either cell viability or cytolytic activity up to 24 h after labeling. Given these parameters, the amount of drug which may be carried to and concentrated in metastatic lesions represents a local concentration of approximately 15 microM. In addition, A-NK cells carrying Zyn-Linked doxorubicin at an equivalent dose of 25 micrograms/kg was therapeutically comparable to a systemic dose of 8 mg/kg (320x more) in the 3LL model of experimental metastasis. These data indicate that A-NK cells bearing Zyn-Linked chemotherapeutic agents represent a unique and feasible method to target chemotherapeutic agents to cancer metastases and that therapeutic doses can be attained without unwanted systemic exposure.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Animals , Antineoplastic Agents/chemistry , Cell Count/drug effects , Doxorubicin/chemistry , Fluorescent Dyes/chemistry , Immunotherapy, Adoptive , Killer Cells, Lymphokine-Activated/cytology , Killer Cells, Lymphokine-Activated/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Survival Analysis , Treatment Outcome , Tumor Cells, Cultured
20.
In Vivo ; 14(1): 265-8, 2000.
Article in English | MEDLINE | ID: mdl-10757085

ABSTRACT

The multicatalytic proteinase complex or proteasome possesses at least 4 distinct proteolytic activities. We have previously reported that the chymotrypsin-like activity of the rat natural killer cell proteasome may play a role in natural killer (NK) cell-mediated cytotoxicity or IL-2 activated NK (A-NK) cell-mediated cytotoxicity. Using a series of novel, Cephalon, Inc, synthetic proteasome inhibitors (CEP-1508, CEP-1612 and CEP-3117) which have been reported to be specific for the chymotrypsin-like activity of the proteasome, we have further investigated the possible role of the proteasome, with emphasis on the chymotryptic activity components, in cell-mediated cytotoxicity. We now report that these compounds can inhibit the rat NK proteasome in a dose dependent manner. Nevertheless, there is only a 50% inhibition of A-NK cell-mediated cytotoxicity. These results confirm and extend our previous results that the proteasome contributes, at least in part, to cell-mediated cytotoxicity. However, as anticipated, since multiple molecular pathways contribute to cell-mediated cytotoxicity, the proteasome contributes only partially to NK cell-mediated cytolytic reactivity. The exact role of the proteasome in NK cell-mediated killing, and whether single or multiple chymotryptic domains function directly or indirectly, remains to be fully determined.


Subject(s)
Cysteine Endopeptidases/drug effects , Lymphocytes/immunology , Multienzyme Complexes/drug effects , Animals , Binding Sites , Cell Survival/drug effects , Chymotrypsin/metabolism , Cysteine Endopeptidases/metabolism , Cytotoxicity, Immunologic/drug effects , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Jurkat Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Multienzyme Complexes/metabolism , Phthalimides/pharmacology , Proteasome Endopeptidase Complex , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...