Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Hum Exp Toxicol ; 34(11): 1096-105, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25636639

ABSTRACT

Naproxen is an anti-inflammatory drug that affects cellular calcium ion (Ca(2+)) homeostasis and viability in different cells. This study explored the effect of naproxen on [Ca(2+)](i) and viability in Madin-Darby canine kidney cells (MDCK) canine renal tubular cells. At concentrations between 50 µM and 300 µM, naproxen induced [Ca(2+)](i) rises in a concentration-dependent manner. This Ca(2+) signal was reduced partly when extracellular Ca(2+) was removed. The Ca(2+) signal was inhibited by a Ca(2+) channel blocker nifedipine but not by store-operated Ca(2+) channel inhibitors (econazole and SKF96365), a protein kinase C (PKC) activator phorbol 12-myristate 13-acetate, and a PKC inhibitor GF109203X. In Ca(2+)-free medium, pretreatment with 2,5-di-tert-butylhydroquinone or thapsigargin, an inhibitor of endoplasmic reticulum Ca(2+) pumps, partly inhibited naproxen-induced Ca(2+) signal. Inhibition of phospholipase C with U73122 did not alter naproxen-evoked [Ca(2+)](i) rises. At concentrations between 15 µM and 30 µM, naproxen killed cells in a concentration-dependent manner, which was not reversed by prechelating cytosolic Ca(2+) with the acetoxymethyl ester of 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl. Annexin V/propidium iodide staining data suggest that naproxen induced apoptosis. Together, in MDCK renal tubular cells, naproxen induced [Ca(2+)](i) rises by inducing Ca(2+) release from multiple stores that included the endoplasmic reticulum and Ca(2+) entry via nifedipine-sensitive Ca(2+) channels. Naproxen induced cell death that involved apoptosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Calcium/metabolism , Madin Darby Canine Kidney Cells/drug effects , Naproxen/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cell Death/drug effects , Dogs , Imidazoles/pharmacology , Indoles/pharmacology , Madin Darby Canine Kidney Cells/metabolism , Maleimides/pharmacology , Nifedipine/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
2.
J Environ Biol ; 30(2): 175-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-20121013

ABSTRACT

This paper presents data on the bioconcentration of heavy metals found in 10 plant species that occur in mangroves and inland ecosystems of India. The average concentration of mercury in the mangrove plants (0.068 microg g(-1)) was 11.3 times that of the inland plants (0.006 microg g(-1); p<0.05). The average concentration of lead in the mangrove plants (19.23 microg g(-1)) was 1.7 times that of the inland plants (11.38 microg g(-1); p<0.05). The mean bioconcentration factors for lead in mangrove plants (2.40 +/- 0.75) were higher than that the inland plants (1.42 +/- 0.15). The factor analysis accounted for 21.55% of the total variance showed accumulation of mercury and lead confirming the polluted nature.


Subject(s)
Metals, Heavy/metabolism , Plants, Medicinal/metabolism , Rhizophoraceae/metabolism , Ecosystem , India
3.
Acta Anaesthesiol Scand ; 53(1): 55-60, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19032555

ABSTRACT

BACKGROUND: It has been proposed that the volatile anesthetic isoflurane induces neuroprotection and that the endogenous opioid peptide dynorphin induces neurocytotoxicity in cells. The levels of dynorphin are often significantly elevated in neuropathophysiological conditions, and dynorphin can directly induce toxicity. However, the neuroprotective effects of isoflurane on dynorphin-induced cytotoxicity are still unclear. METHODS: In order to determine the effect of isoflurane on dynorphin-induced cytotoxicity in neuronal cells, we have designed a device wherein cultured human neuroblastoma SH-SY5Y cells can be exposed to isoflurane. Fully differentiated SH-SY5Y cells were obtained by treating the cells with retinoic acid for 6 days. We examined SH-SY5Y cell survival, apoptosis, and antiapoptotic protein expression by cell viability, terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling stain, and Western blot analysis, respectively. RESULTS: After 16 h of dynorphin (10 microM) treatment, the SH-SY5Y cells showed significant cytotoxicity, apoptosis, and downregulation of the antiapoptotic Bcl-2 protein expression. These effects of dynorphin were significantly inhibited by isoflurane exposure for 32 h [pretreatment for 16 h and posttreatment (after dynorphin treatment) for 16 h]. CONCLUSION: Thus, our results suggest that isoflurane exerts neuroprotective effects in the case of dynorphin-induced pathophysiological disruption.


Subject(s)
Cell Differentiation , Down-Regulation/drug effects , Dynorphins/toxicity , Isoflurane/pharmacology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Humans
4.
Food Chem Toxicol ; 46(6): 1925-32, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18321630

ABSTRACT

The aim of this study was to examine levels of PBDEs in breast milk associated with seafood consumptions of Taiwanese mothers. Our participants were selected from healthy women recruited between December 2000 and November 2001 from a medical center in central Taiwan. The congeners of PBDEs in 20 milk samples were analyzed by a gas chromatograph with a high resolution mass detector. The mean level of BDE47 in breast milk from mothers with pre-pregnant BMI <22.0kg/m2 had a significantly higher magnitude compared to those with pre-pregnant BMI > or = 22.0kg/m2 (1.59 vs. 0.995ng/g lipid, p=0.041). We did not find significant correlations between PBDEs exposure levels and women's age, parity, blood pressure, annual household income, and education level. Women who ate more fish and meat did not show significantly higher PBDE levels than those who ate less, but a significant difference in PBDE levels was demonstrated between the higher (2.15ng/g lipid) and lower (3.98ng/g lipid) shellfish consuming subjects (p=0.002) after an adjustment for the confounders. The ratios of PCB153/BDE47, PCB153/BDE153, and PCB153/PBDEs were significantly correlated with frequent consumption of fish and shellfish. The PCB153/BDE153 ratio was not associated with the other dietary habits (i.e. meat). The ratios of PCB153/PBDEs may therefore be a new indicator for exposure as a result of seafood consumption.


Subject(s)
Feeding Behavior/physiology , Milk, Human/chemistry , Phenyl Ethers/analysis , Polybrominated Biphenyls/analysis , Adult , Age Factors , Blood Pressure/physiology , Cohort Studies , Dioxins/analysis , Female , Halogenated Diphenyl Ethers , Humans , Parity , Pregnancy , Seafood , Shellfish , Socioeconomic Factors , Taiwan/epidemiology
5.
Cell Mol Life Sci ; 61(11): 1372-83, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15170515

ABSTRACT

We applied a robust combinatorial (multi-test) approach to microarray data to identify genes consistently up- or down-regulated in head and neck squamous cell carcinoma (HNSCC). RNA was extracted from 22 paired samples of HNSCC and normal tissue from the same donors and hybridized to the Affymetrix U95A chip. Forty-two differentially expressed probe sets (representing 38 genes and one expressed sequence tag) satisfied all statistical tests of significance and were selected for further validation. Selected probe sets were validated by hierarchical clustering, multiple probe set concordance, and target-subunit agreement. In addition, real-time PCR analysis of 8 representative (randomly selected from 38) genes performed on both microarray-tested and independently obtained samples correlated well with the microarray data. The genes identified and validated by this method were in comparatively good agreement with other rigorous HNSCC microarray studies. From this study, we conclude that combinatorial analysis of microarray data is a promising technique for identifying differentially expressed genes with few false positives.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Algorithms , Carcinoma, Squamous Cell/genetics , Cluster Analysis , Gene Expression Profiling/standards , Humans , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Array Sequence Analysis/standards , Statistics as Topic
6.
J Exp Clin Cancer Res ; 20(2): 175-82, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11484971

ABSTRACT

Epstein Barr Virus (EBV) associated lymphoproliferative disorders (LPD) express EBV latent antigens that are also expressed on normal B-cells transformed with EBV. This could potentially be exploited to develop immunotherapeutic strategies for LPD and other EBV associated malignancies. To this end we investigated the capacity of human monocyte derived dendritic cells (DC) pulsed with lysate from autologous EBV transformed B-cell lymphoblastoid cell (BCL) lysate to elicit an in vitro antitumor response. BCL lysate pulsed DC generate BCL specific cytotoxic lymphocytes, as lymphocytes primed with such DCs induce cytolysis of autologous (>60%) but not allogeneic BCL (<5%). In addition, lymphocytes primed with BCL lysate pulsed DC secrete gamma-IFN (3176 pg/ml). Whereas gamma-IFN production was markedly reduced (>99%) when BCL specific T-cells were stimulated by BCL lysate pulsed DC in the presence of blocking antibodies to HLA-DR, DP and DQ, use of antibodies to MHC class-I resulted in only a minimal reduction in gamma-IFN production (17%). These studies demonstrate that BCL lysate pulsed DC elicit a predominantly BCL specific, MHC class-Il restricted T cell response. This suggests that vaccination with autologous BCL lysate pulsed DC may represent a viable immunotherapeutic approach for the treatment of LPD.


Subject(s)
B-Lymphocytes/immunology , Cell Transformation, Viral/immunology , Dendritic Cells/immunology , Herpesvirus 4, Human/physiology , Histocompatibility Antigens Class II/immunology , T-Lymphocytes, Cytotoxic/immunology , Cells, Cultured , Cytokines/biosynthesis , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation , Monocytes/immunology
7.
Int J Cancer ; 92(4): 589-99, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11304696

ABSTRACT

To determine the possible functional significance of CD40 expression on human non-small cell lung carcinomas and to assess the potential of CD40 as a therapeutic target, 18 lung tumor cell lines were established from biopsy tissues and were monitored for phenotypic changes on the cell surface and alterations in tumor cell proliferation after the ligation of CD40 with a trimeric fusion protein complex of CD40 ligand (CD40Lt). CD40 cross-linking resulted in up to a 6-fold increase in the surface expression of major histocompatibility complex (MHC) class I, Fas and intracellular adhesion molecule (ICAM)-1 in a subset of tumors expressing the highest levels of CD40. Suppression of tumor proliferation was seen after the ligation of CD40 on CD40Lt-responsive cell lines. The suppression was dose dependent, reversible and resulted from a delay of the tumor cells entering S-phase. No change in the cell phenotype or in proliferation were observed in CD40-negative tumors or in tumors expressing moderate-to-low levels of CD40 after incubation with CD40Lt. CD40-negative tumors transfected with the CD40 gene expressed high levels of CD40 on their surface, but were also unresponsive to CD40Lt cross-linking of CD40. Our data establish that CD40 is required (but not sufficient) for transducing a signal that results in phenotypic changes in human lung tumors and suppression in their proliferation. We conclude that CD40 on non-small cell lung tumors may represent a potential therapeutic target, but only on a subset of the CD40+ tumors.


Subject(s)
CD40 Antigens/biosynthesis , CD40 Ligand/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Lung Neoplasms/metabolism , Major Histocompatibility Complex , fas Receptor/biosynthesis , Apoptosis , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cell Cycle , Cell Division , Coloring Agents/pharmacology , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunohistochemistry , Phenotype , Recombinant Fusion Proteins/metabolism , S Phase , Signal Transduction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Transfection , Tumor Cells, Cultured
8.
J Immunother ; 24(1): 37-45, 2001.
Article in English | MEDLINE | ID: mdl-11211147

ABSTRACT

The human tumor microenvironment includes a mixture of tumor cells, inflammatory cells, fibroblasts, and endothelial cells, all of which are tethered to an extracellular matrix. It has been difficult to study the dynamic interactions of these cells in human tumors in situ for obvious ethical and logistical considerations that prohibit experimental manipulations of tumors while still in patients. Fresh tissue from human lung tumor biopsy implanted into SCID mice was shown to remain viable, and the histologic appearance of the tumor microenvironment was maintained in the tumor xenografts for at least 3 months. In this study, the authors established that the inflammatory cells within human tumor xenografts can suppress tumor growth, and that this suppression is a result, in part, of endogenously produced interleukin-12 (IL-12) because IL-12 neutralizing antibodies enhance the growth of the tumor xenografts. The tumor-inhibitory activity of the inflammatory leukocytes is also enhanced by the local and sustained release of human recombinant IL-12 into the tumor microenvironment from cytokine-loaded biodegradable microspheres. Neither the anti-IL-12 neutralizing antibody nor the delivery of exogenous IL-12 from microspheres had any effect on tumor xenografts in the absence of the inflammatory leukocytes. In conclusion, the inflammatory cells within the tumor microenvironment of human lung tumor xenografts are functional and can suppress tumor growth, and the dynamic effects of the inflammatory cells can be modulated by exogenous cytokines.


Subject(s)
Adjuvants, Immunologic/physiology , Interleukin-12/physiology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Transplantation, Heterologous/immunology , Adjuvants, Immunologic/genetics , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/genetics , Cell Division/immunology , Growth Inhibitors/genetics , Growth Inhibitors/pharmacology , Humans , Inflammation/immunology , Inflammation/pathology , Interleukin-12/genetics , Interleukin-12/immunology , Lung/growth & development , Lung/immunology , Lung Neoplasms/prevention & control , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, SCID , Recombinant Proteins/pharmacology , Transplantation, Heterologous/pathology
9.
J Chromatogr Sci ; 39(1): 7-11, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11206914

ABSTRACT

A rapid, sensitive, and accurate stability-indicating high-performance liquid chromatographic assay method for determining the degradation of carprofen (CPF) is developed and validated under acidic, basic, or photo-irradiated conditions. The analysis is monitored with a Cosmosil 5C18-AR column using a mobile phase of CH3CN-H2O-AcOH (50:49:1, v/v/v) at 260 nm. The developed method satisfies the system suitability criteria, peak integrity, and resolution among the parent drug and its degradation products. The results indicate that the established assay method shows good selectivity and specificity suitable for stability measurements of CPF. CPF is found to be more sensitive to exposure to light and in acidic conditions, but it is stable in a basic medium. The kinetic study of the photodegradation of CPF follows an apparent first-order reaction in a variety of solvents. The solvent effects on the rates of degradation are in the decreasing order of chloroform > dichloromethane > methanol > ethanol > 2-propanol, which is irrelevant to the dielectric constant epsilon. However, the hydrogen-donating ability of the solvents is essential to the photochemical decomposition of CPF. A plot of log k versus the Kirkwood function exhibits a linear relationship in aqueous ethanolic solutions, which implies that degradation proceeds via an ionic mechanism.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/analysis , Carbazoles/analysis , Chromatography, High Pressure Liquid/methods , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Carbazoles/chemistry , Drug Stability , Kinetics , Reproducibility of Results , Sensitivity and Specificity , Solvents , Spectrophotometry, Ultraviolet
10.
Laryngoscope ; 111(10): 1842-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11801955

ABSTRACT

OBJECTIVE/HYPOTHESIS: To identify the significance of molecular markers in determining the risk of recurrence and distant metastases in nasopharyngeal carcinoma. STUDY DESIGN: In this retrospective case study, we evaluated archival nasopharyngeal carcinoma specimens for patterns of expression of E-cadherin, beta-catenin, c-erb-B2, and Ki-67, which have been demonstrated to be important in other tumors. METHODS: Fifty-four cases of nasopharyngeal carcinoma were identified, with a maximum follow-up of 13 years. The histopathological sections were stained using an automated immunohistochemical stainer (NexES, Ventana Medical Systems, Tucson, AZ) for E-cadherin (Zymed Laboratories [San Francisco, CA] and Transduction Laboratories [Lexington, KY] clones), beta-catenin (Zymed), c-erb-B2 (Ventana Medical Systems), and Ki-67 (Novocastra, Burlingame, CA). The numbers of positively staining cells were scored as follows: 0%, 1% to 33%, 34% to 66%, or greater than 67%. RESULTS: E-cadherin (Zymed) stained positively in only one case. The Transduction Laboratories clone demonstrated a spectrum of staining in all cases, from complete to disrupted to no identifiable membranous staining. The staining was consistently absent at the advancing tumor border, regardless of stage. The loss of beta-catenin expression did not correlate with that of E-cadherin or with clinical outcomes. No staining was identified for c-erb-B2. Ki-67 staining was variable and did not correlate with clinical outcomes. CONCLUSIONS: Altered expression or loss of E-cadherin, or both, may result in loss of function, particularly at the infiltrating edge, with resultant loss of cell polarity, cell migration, and eventual metastasis. The interpretation of E-cadherin staining depends on antibody source. In contrast to recent studies, beta-catenin expression is not altered and c-erb-B2 expression not identified, suggesting that these markers are not important in the prognosis of nasopharyngeal carcinoma.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecules/analysis , Nasopharyngeal Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Trans-Activators , Adult , Aged , Cadherins/analysis , Cytoskeletal Proteins/analysis , Disease-Free Survival , Female , Follow-Up Studies , Humans , Ki-67 Antigen/analysis , Male , Middle Aged , Nasopharynx/pathology , Neoplasm Invasiveness , Neoplasm Staging , Receptor, ErbB-2/analysis , Retrospective Studies , beta Catenin
12.
J Immunother ; 23(3): 362-8, 2000.
Article in English | MEDLINE | ID: mdl-10838665

ABSTRACT

A monoclonal antibody specific for the human analog of the murine T-cell activation molecule 4-1BB was generated and is shown here to react selectively with activated human CD4+ and CD8+ T lymphocytes. Treatment of these T cells in a one-way mixed lymphocyte culture with the anti-h4-1BB antibody enhanced the cell proliferation of the allostimulated lymphocytes. Previous studies in the mouse have shown that treatment of tumor-bearing mice with antibodies to 4-1BB augments anti-tumor immunity that is mediated by both CD4+ and CD8+ T cells. The authors consider the possibility that a similar approach may be efficacious for human cancer immunotherapy. This question was addressed by evaluating the effect of an anti-h4-1BB monoclonal antibody on human lymphocyte-mediated suppression of a human tumor xenograft in SCID mice. Mice treated with a control antibody and co-injected with the tumor and peripheral blood lymphocytes exhibited a lymphocyte dose-dependent suppression of tumor growth. In mice treated with the anti-h4-1BB antibody, the lymphocyte-mediated tumor suppression was completely eliminated and tumors grew progressively (as was observed in mice inoculated with tumors without lymphocytes). This monoclonal antibody specific for anti-h4-1BB, which augments the proliferation of allostimulated cells in vitro, blocks T-cell anti-tumor activity in vivo. These results suggest that although 4-1BB plays a role in the human peripheral blood lymphocyte-mediated suppression of tumor growth, antibodies to this molecule on human cells fail to stimulate anti-tumor activity, as was observed in tumor-bearing mice treated with an antibody to murine 4-1BB.


Subject(s)
Lung Neoplasms/immunology , Lymphocytes/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Severe Combined Immunodeficiency/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD , CD4 Antigens/immunology , CD8 Antigens/immunology , Dendritic Cells/immunology , Humans , Isoantigens/immunology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 9
13.
Cancer Immunol Immunother ; 49(2): 101-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10823420

ABSTRACT

PURPOSE: The poor prognosis associated with lung cancer is related to the high incidence of regional and distant metastasis. There is a crucial need to identify parameters that can predict a tendancy to metastatic spread to allow better prognostic evaluation and therapeutic approach. METHODS: Using flow cytometry we evaluated 18 human lung cancer cell lines for the expression of different surface markers on lung cancers suggested to be possible prognostic parameters, including epidermal growth factor receptor (EGFR), intercellular adhesion molecule 1 (ICAM-1), Fas and CD40. RESULTS: No correlation was found between tumor prognosis and EGFR, ICAM-1 or Fas. However, a statistically significant correlation was found between the surface expression of CD40 and the metastatic spread of the tumor. In this study, 14 of 18 lung cancer cell lines (78%) expressed CD40 on their surface. All of the 4 tumors that were CD40-negative, were stage I tumors, without any evidence of regional or distant metastasis. Of the 14 tumors that expressed CD40, all but 1 (93%) had either nodal or systemic metastasis at the time of diagnosis. Patients whose tumors were CD40-negative showed a significantly better N stage, overall stage at presentation and survival than those patients with CD40-positive patients. No significant differences between the two groups were observed in tumor size, gender, age, histology, differentiation or preoperative therapy. CONCLUSIONS: These results suggest that CD40 expression on lung cancer may play a role in metastatic spread, and also may serve as a prognostic marker and an indicator of advanced disease.


Subject(s)
CD40 Antigens/analysis , Lung Neoplasms/chemistry , Neoplasm Metastasis , ErbB Receptors/analysis , Humans , Intercellular Adhesion Molecule-1/analysis , Lung Neoplasms/pathology , Prognosis , Tumor Cells, Cultured
14.
Cancer Res ; 60(24): 6942-9, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11156394

ABSTRACT

Beta1 integrins, expressed on the cell surface of human non-small cell lung carcinomas, are used here as a target for the selective delivery of anti-cancer drug-loaded liposomes. Fab' fragments of a monoclonal antibody specific for human beta1 integrins were conjugated to sterically stabilized liposomes. Confocal microscopy of beta1 integrin-positive lung tumor cells incubated with fluorescently labeled anti-beta1 Fab immunoliposomes revealed a tumor-specific binding and efficient internalization of the liposomes into the tumor cells. The ability of these liposomes to deliver cytotoxic drugs to the tumor and kill these cells was demonstrated in vitro by incubating tumor cells with doxorubicin-loaded anti-beta1 Fab' immunoliposomes. The drug-loaded immunoliposomes were >30-fold more cytotoxic to the tumor cells than drug-loaded liposomes without antibody, nonspecific Fab' control immunoliposomes with drug or immunoliposomes without drug. The therapeutic efficacy of doxorubicin-loaded immunoliposomes was also evaluated in a metastatic human lung tumor xenograft/severe combined immunodeficient (SCID) mouse model. SCID mice that received i.v. injections of human lung tumor cells developed primary tumor nodules in the lung that subsequently metastasized to the liver and adrenal gland. Treatment of SCID mice bearing established lung tumor xenografts with doxorubicin-loaded anti-beta1 Fab immunoliposomes resulted in a significant suppression of tumor growth (monitored periodically by quantifying serum levels of a tumor marker), whereas tumors grew progressively in mice treated with control formulations. In addition to suppressing the growth of the primary lung tumor nodules, the immunoliposomes prevented the metastatic spread of the tumor to the liver and adrenal glands and increased the median survival time of the tumor-bearing mice. We conclude that Fab' immunoliposomes directed to tumor-associated integrins represent a potentially viable approach clinically for the selective delivery of drugs to solid tumors and may be useful in preventing the metastatic spread of lung cancer.


Subject(s)
Doxorubicin/pharmacology , Liposomes/immunology , Liposomes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Adrenal Gland Neoplasms/secondary , Animals , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Integrins/immunology , Liposomes/therapeutic use , Liver Neoplasms/secondary , Male , Mice , Mice, SCID , Microscopy, Confocal , Neoplasm Transplantation , Protein Binding , Time Factors , Tumor Cells, Cultured
15.
Head Neck ; 22(1): 57-63, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10585606

ABSTRACT

BACKGROUND: The role of cytokines in tumor regression is now well established. The major limitation for the clinical use of cytokines is the lack of a simple and effective protocol for the local and sustained delivery of cytokines to the tumor milieu. This study reports suppression of human head and neck squamous cell carcinoma (HNSCC) by human peripheral blood lymphocytes (HuPBL) following local, sustained delivery of interleukin-12 (IL-12) to tumors with biodegradable microspheres in a human/SCID mouse chimeric model. Materials and Methods Nondisrupted biopsy pieces (120 mg) of primary HNSCC were implanted s.c. into severe combined immunodeficient (SCID) mice and were expanded by serial passage in mice. Tumors were then titrated with different doses of allogeneic HuPBL by coengraftment of tumor pieces and HuPBL into the subcutis of SCID mice to determine whether the HuPBL possessed antitumor activity (the SCID/Winn model). The lymphocyte subsets that were responsible for the suppression of tumor engraftment were identified by selective depletion of the CD4+, CD8+, and CD56+ cells from the HuPBL prior to engraftment into mice. Attempts were then made to augment the antitumor activity of the HuPBL either by repeated intralesional bolus injections of recombinant human IL-12 (0.5 microg x 10 doses) or with a single dose of IL-12-loaded microspheres ( approximately 1.65 microg IL-12/mg microspheres, 2 mg microspheres/mouse). RESULTS: Successful engraftment of HNSCC was observed in 12 of 19 different patient samples. Normal histological architecture of tumor was maintained up to four serial passages in the SCID mice. After the first tumor engraftment, but not in subsequent passages, human immunoglobulin produced by plasma cells present in the tumor infiltrating lymphocyte population was detected in the mouse sera. Allogeneic human PBL displayed antitumor cytotoxic activity in a cell dose-dependent fashion when coengrafted with the tumors passaged in SCID mice. Lymphocyte subset depletion studies established that tumor suppression was dependent on both the CD8+ T lymphocytes and the CD56+ natural killer cells. Treatment of tumors with a single intralesional injection of IL-12-loaded microspheres was highly effective, resulting in the complete suppression of tumor engraftment in 50% of the mice. In contrast, treatment of tumors with repeated bolus IL-12 injections suppressed tumor engraftment only transiently and did not result in complete tumor rejection in any of the mice. CONCLUSION: The coengraftment of HNSCC and allogeneic lymphocytes into SCID mice provides a viable model with which to evaluate immunotherapeutic strategies for human cancer. The use of biodegradable microspheres for local sustained delivery of cytokines to augment lymphocyte mediated antitumor immunity within the tumor microenvironment provides a safer and simpler alternative to current cytokine immunotherapy protocols.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Interleukin-12/administration & dosage , Lymphocytes, Tumor-Infiltrating/drug effects , Animals , Biodegradation, Environmental , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Carriers , Flow Cytometry , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Injections, Intralesional , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, SCID , Microspheres , Reference Values , Sensitivity and Specificity , Transplantation, Heterologous
16.
Cancer Immunol Immunother ; 46(1): 21-4, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9520288

ABSTRACT

A novel biodegradable poly(lactic acid) microsphere formulation was evaluated for in vivo cytokine immunotherapy of cancer in a human tumor xenograft/ severe combined immunodeficiency (SCID) mouse model. Co-injection of interleukin-2 (IL-2)-loaded microspheres with tumor cells into a subcutaneous site resulted in the complete suppression of tumor engraftment in 80% of animals. In contrast, bovine-serum-albumin(BSA)-loaded particles or bolus injections of poly(ethylene glycol)/IL-2 were ineffective in preventing tumor growth. The antitumor effect of IL-2 released by the microspheres was shown to be mediated by the mouse natural killer cells. This is the first evidence that the rejection of human tumor xenografts can be provoked by the sustained in vivo delivery of IL-2 from biodegradable microspheres. The use of poly(lactic acid) microspheres to deliver cytokines to the tumor environment could provide a safer and simpler alternative to gene therapy protocols in the treatment of cancer.


Subject(s)
Interleukin-2/administration & dosage , Killer Cells, Natural/drug effects , Neoplasms, Experimental/drug therapy , Animals , Carcinoma, Squamous Cell/drug therapy , Delayed-Action Preparations , Drug Delivery Systems , Humans , Interleukin-2/chemistry , Lactic Acid , Lung Neoplasms/drug therapy , Mice , Mice, SCID , Microspheres , Polyesters , Polymers , Tumor Cells, Cultured
17.
Cancer Res ; 57(14): 2937-42, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9230205

ABSTRACT

Here, it is established that human peripheral blood lymphocytes (HuPBLs), injected s.c. with a human lung tumor into severe combined immunodeficient (SCID) mice, engraft and display antitumor cytotoxic activity. Initial studies used HuPBLs from normal donors and an allogeneic tumor cell line derived from biopsy tissue of a patient with a squamous cell carcinoma of the lung. Evidence of HuPBL antitumor activity was revealed by a cell dose-dependent suppression of the tumor xenograft. Tumor suppression was shown to be dependent upon both CD8+ T cells and CD56+ natural killer cells in the donor HuPBLs. By titrating the antitumor activity of HuPBLs in SCID mice with and without cytokines, it was established that interleukin (IL)-12 enhanced the HuPBL-mediated tumor suppression and that IL-2 had a synergistic effect upon the IL-12 enhancement of cytotoxicity. Subsequent studies revealed that a lung cancer patient's PBLs also suppress the growth of the patient's (autologous) tumor when coinjected s.c. with the tumor cells into SCID mice. The patient's antitumor immunity was shown to be mediated by CD8+ T cells and CD56+ natural killer cells. The data presented here indicate that the s.c. coengraftment of HuPBLs and tumor into SCID mice represents a viable model with which to study (and to periodically monitor) patients' immune responses to their tumors for extended periods of time and suggest that this SCID/Winn assay could be used to evaluate novel immunotherapeutic approaches, such as bolus injections of cytokines, cytokine gene therapy, or vaccination strategies for the treatment of human cancer.


Subject(s)
Lymphocytes/immunology , Neoplasms/immunology , Animals , Antibodies, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Interleukin-12/pharmacology , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Mice , Mice, SCID , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Cancer J Sci Am ; 2(5): 291-300, 1996.
Article in English | MEDLINE | ID: mdl-9166547

ABSTRACT

PURPOSE: We have studied the growth and metastatic potential of surgical specimens of breast carcinomas engrafted into the large abdominal (gonadal) fat pad of severe combined immunodeficient (SCID) mice. We present results of this study, details of the implantation protocol and histologic characterization of several of the tumor xenografts. MATERIALS AND METHODS: We evaluated the growth within SCID mice of 48 breast carcinoma specimens derived from 46 patients (45 primary breast cancers or local recurrences and 3 regional metastatic lymph nodes) obtained from resected tissues at this Institute over a 3-year period. The growth of each transplant was assessed by histologic examination of the xenografts at various times after implantation or upon passage into additional mice. RESULTS: We observed that placement of human breast tumors within the gonadal fat pad could result in tumors that grew either rapidly, slowly, or not at all. Of 48 tumors studied, 12 (25%), including one of the three lymph node-derived tumors, grew rapidly enough within some or all of the implanted mice (i.e., the tumors reached a diameter of 2-3 cm within 2-6 months) to allow repeated passage. Metastatic spread to the SCID mouse lung, liver, and/or diaphragm and other sites was observed with the xenografts derived from 8 of these 12 rapidly growing tumors. Tumors in a second category often took from 6 months to over 1 year to only double or triple in size. This slow-growth group consisted of 25 patients' tumors (53%), including the remaining two metastatic lymph node-derived tumors. These xenografts would usually maintain a slow growth rate even upon later passage into new animals. A third category consisted ofpatients' tumors (23%) that failed to grow at all (i.e., no evidence of tumor growth in any of the mice implanted), as discerned by histologic evaluation at various times after implantation. Histologic examination of tumor xenografts and metastatic tumors revealed considerable variation in histopathology among the different patients' tumors. DISCUSSION: Further examination of the heterogeneous properties of primary human breast carcinomas within SCID mice may provide a simple yet valuable new approach for the long-term study of human breast cancer biology. Importantly, use of the protocol described here can often permit the isolation of substantial quantities of human breast cancer cells for biochemical and molecular analyses. The ability to passage patients' breast tumors into large numbers of mice will permit the preclinical testing of new therapies for the treatment and prevention of this disease.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Carcinoma, Lobular/secondary , Severe Combined Immunodeficiency/pathology , Animals , Biomarkers, Tumor/blood , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/surgery , Carcinoma, Lobular/surgery , Female , Humans , Lymphatic Metastasis , Mice , Mice, SCID , Transplantation, Heterologous , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/transplantation
19.
Cancer Res ; 56(16): 3743-6, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8706018

ABSTRACT

Liposomes containing polyethylene glycol-derivatized phospholipids are able to evade the reticuloendothelial system and thereby remain in circulation for prolonged periods. We report here that doxorubicin encapsulated in these sterically stabilized liposomes (S-DOX) suppresses the growth of established human lung tumor xenografts in severe combined immunodeficient (SCID) mice and inhibits the spontaneous metastases of these tumors. The enhanced therapeutic efficacy of S-DOX compared to free doxorubicin was demonstrated in two independent human/mouse models. In the first model, S-DOX inhibited the growth of a human non-small cell lung tumor xenograft established orthotopically in the lungs of SCID mice. Treatment of these mice with S-DOX, but not with free drug, suppressed the growth of the tumor in the lung, prevented metastasis from the lung, and enhanced survival percentage. In another model, the human lung tumor is engrafted into gonadal fat pad of SCID mice. Human tumor xenografts grow floridly in this site of engraftment, and the tumor spreads from this primary site into the peritoneal cavity and subsequently reaches the liver and lung. In this model, free drug suppressed the growth of the primary tumor but had no effect upon the subsequent spread of the tumor into the peritoneal cavity, liver, and lung. In contrast, treatment of the tumor-bearing mice with S-DOX (but not with doxorubicin in conventional liposomes) suppressed the tumor spread to the peritoneal cavity, completely arrested metastasis to the liver and lung, and suppressed the growth of the primary tumor xenograft. This report provides the first evidence that antitumor drugs delivered by sterically stabilized liposomes can arrest the metastasis of human tumor xenografts.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Lung Neoplasms/drug therapy , Animals , Drug Carriers , Humans , Liposomes , Mice , Mice, Inbred C3H , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Transplantation, Heterologous
20.
J Immunol ; 156(5): 1908-15, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8596043

ABSTRACT

We report here the placement of nondisrupted 1-mm3 pieces of fresh human lung tumor biopsy tissue into the subcutis of severe combined immunodeficient (SCID) mice results in the engraftment of tumor-infiltrating leukocytes (TIL) in all but 5 of 148 mice inoculated with 39 different biopsy tissue specimens. In mice coengrafted with tumor and TIL the normal histologic architecture of the tumor and TIL interface was maintained for up to 22 wk. The TIL in the xenograft were shown to divide and were maintained exclusively at the site of tumor inoculation. It is established here that plasma cells in the TIL population produce Abs that react in western blots with tumor cell lysates. These Abs were shown to react with high and low m.w. proteins derived from both the membrane and cytosolic fractions of tumor cell lysates. The production of human Ig was found to be T cell dependent, and immunohistochemistry and in situ hybridization of DNA, using a human-specific cDNA probe, established the human identity of the tumor and TIL. High levels of human Ig in the sera of mice inoculated with tumor biopsy tissue are associated with the growth arrest of adenocarcinoma xenografts. Our results establish the co-engraftment of human tumors and TIL into SCID mice as new animal model with which to evaluate TIL function and novel therapeutic strategies that are designed to augment TIL anti-tumor activity.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Graft Survival/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/transplantation , Transplantation, Heterologous/immunology , Animals , Cell Division/immunology , Humans , Lung Transplantation/immunology , Lung Transplantation/pathology , Lymphocyte Transfusion , Mice , Mice, SCID , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transplantation, Heterologous/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...