Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 50(6): 285-92, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11570581

ABSTRACT

In this study, we report on novel alterations found in rat intracranial (i.c.) tumor-infiltrating T lymphocytes (TIL) that are indicative of T cell defects and death. FACS analysis showed that the cytotoxic T cells (CTL) infiltrating rat T9.F gliomas were CD3epsilon+, alphabetaTCR+, CD8alpha+, but CD8beta-. These lymphocytes also stained positive for the B cell-specific marker, CD45RA, as well as Annexin-V, signifying apoptotic changes. Functional and biochemical analyses were performed to assess whether the aberrant phenotype was linked to other defects. When CD8alpha+ TIL were purified and stimulated in vitro, their proliferative capacity was markedly diminished in comparison with CD3+CD8alpha+CD8beta+ T cells isolated from the spleens of naive, non tumor-bearing rats. Furthermore, the mean fluorescence intensity of surface CD3epsilon was dramatically reduced in the CD3+CD8alpha+CD8beta- TIL population as compared with CD3-CD8alpha+CD8beta+ TIL from the same tumor-bearing animal. Biochemical studies revealed that the expression of TCRzeta and LAT were reduced in lysates generated from CD8alpha-purified TIL with respect to CD8alpha-purified T cells from naive spleen. We believe that these degenerative changes are reflective of chronic T cell receptor ligation, because in vitro culture of rat splenocytes or purified T cells with ConA or anti-CD3 mAb induced the same alterations. In vitro, the downregulation of CD8beta could be inhibited by the caspase inhibitor, z-VAD. These results suggest that the aberrant CTL phenotype found in the TIL of glioma-bearing rats may be novel signals for their impending death and degenerating anti-tumor immune function.


Subject(s)
Apoptosis/immunology , Brain Neoplasms/immunology , Glioma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Annexin A5/biosynthesis , CD8 Antigens/biosynthesis , Down-Regulation/immunology , Female , Immunophenotyping , Leukocyte Common Antigens/biosynthesis , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/physiology , Rats , Rats, Inbred F344 , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/physiology
2.
Altern Ther Health Med ; 7(3): 79-91, 2001.
Article in English | MEDLINE | ID: mdl-11347287

ABSTRACT

CONTEXT: It has been suggested that the consumption of natural "whole foods" rich in macronutrients has many healthful benefits for those who otherwise ingest a normal, nonvegetarian diet. One example is dietary supplements derived from Chlorella pyrenoidosa, a unicellular fresh water green alga rich in proteins, vitamins, and minerals. OBJECTIVE: To find evidence of the potential of chlorella dietary supplements to relieve signs and symptoms, improve quality of life, and normalize body functions in people with chronic illnesses, specifically fibromyalgia, hypertension, and ulcerative colitis. DESIGN: Double-blind, placebo-controlled, randomized clinical trials. SETTING: Virginia Commonwealth University's Medical College of Virginia. PATIENTS: Fifty-five subjects with fibromyalgia, 33 with hypertension, and 9 with ulcerative colitis. INTERVENTION: Subjects consumed 10 g of pure chlorella in tablet form and 100 mL of a liquid containing an extract of chlorella each day for 2 or 3 months. MAIN OUTCOME MEASURES: For fibromyalgia patients, assessments of pain and overall quality of life. For hypertensive patients, measurements of sitting diastolic blood pressure and serum lipid levels. For patients with ulcerative colitis, determination of state of disease using the Disease Activity Index. RESULTS: Daily dietary supplementation with chlorella may reduce high blood pressure, lower serum cholesterol levels, accelerate wound healing, and enhance immune functions. CONCLUSIONS: The potential of chlorella to relieve symptoms, improve quality of life, and normalize body functions in patients with fibromyalgia, hypertension, or ulcerative colitis suggests that larger, more comprehensive clinical trials of chlorella are warranted.


Subject(s)
Chlorella , Colitis, Ulcerative/prevention & control , Dietary Supplements , Fibromyalgia/prevention & control , Hypertension/prevention & control , Humans , Randomized Controlled Trials as Topic
3.
J Immunol ; 166(1): 121-9, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11123284

ABSTRACT

Previously, we reported that IL-6 transduction attenuates tumor formation of a rat T9 glioma clone (termed T9.F). This study focuses on the mechanisms of the antitumor response elicited by IL-6 and the generation of glioma immunity. Ten days post s.c. inoculation of T9. F- or IL-6-secreting T9.F cells (T9.F/IL6/hi), tumor nodules were removed and their leukocytic infiltrate was analyzed by FACS with Ab markers for T cells, B cells, granulocytes, and monocytes. T9. F/IL6/hi tumors showed a marked increase in granulocytes as compared with parental T9.F tumors, and histological examination revealed that the granulocytes were neutrophils. Animals made neutropenic failed to reject T9.F/IL6/hi tumors. FACS analysis of 17-day T9. F/IL6/hi regressing tumors and T9.F progressing tumors did not reveal any significant differences in the leukocytic infiltrates. Tumor-specific effector cells were detected in the spleens harvested from animals bearing 17-day, regressing, T9.F/IL6/hi tumors. In vitro, these effector cells lysed T9.F cells, proliferated in response to T9.F stimulator cells, and produced Th1 cytokines (IL-2 and IFN-gamma) but not the Th2 cytokine, IL-4, when cocultured with T9.F stimulator cells. Rats that had rejected s.c. T9.F/IL6/hi tumors displayed a delayed-type hypersensitivity response when injected with viable T9.F cells in the contralateral flank. Passive transfer of spleen cells from these animals transferred glioma immunity to naive recipients and depletion of CD3(+) T cells, before transfer, completely abolished immunity, whereas depletion of CD8(+) T cells had moderate inhibitory effects on the transfer of immunity.


Subject(s)
Glioma/immunology , Glioma/metabolism , Interleukin-6/metabolism , Neutrophils/immunology , Adoptive Transfer , Animals , Clone Cells , Coculture Techniques , Female , Glioma/prevention & control , Graft Rejection/immunology , Immunity, Cellular , Immunity, Innate , Immunologic Memory , Injections, Intraventricular , Injections, Subcutaneous , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Spleen/cytology , Spleen/transplantation , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/transplantation
4.
Phytother Res ; 14(3): 167-73, 2000 May.
Article in English | MEDLINE | ID: mdl-10815009

ABSTRACT

Fibromyalgia syndrome is a common, chronic musculoskeletal disorder of unknown aetiology. While available therapy is often disappointing, most patients can be helped with a combination of medication, exercise and maintenance of a regular sleep schedule. The objective of the present study was to determine if adding nutritional supplements derived from the unicellular green alga, Chlorella pyrenoidosa, produced any improvements in the clinical and functional status in patients with moderately severe symptoms of fibromyalgia syndrome. Eligible patients had 2+ palpable tenderness at 11 or more of 18 defined tender points and had a tender point index (TPI) of at least 22. Each day for 2 months, participants consumed two commercially available Chlorella-based products, 10 g of 'Sun Chlorella' tablets and 100 mL of liquid 'Wakasa Gold'. Any amelioration of symptoms was validated and quantified using semi-objective and subjective outcome measures systematically administered at clinic visits on days 0, 30 and 60 of the diet therapy. Eighteen of the 20 patients enrolled completed the 2 month trial. The average TPI for the group which at onset was 32, decreased to a mean of 25 after 2 months. This decrease was statistically significant (p = 0.01), representing a 22% decrease in pain intensity. Blood samples taken on each occasion indicated no significant alterations in serum chemistries, formed elements, and circulating lymphocyte subsets. Compilations of the results of patient interviews and self-assessment questionnaires revealed that seven patients felt that the dietary supplement had improved their fibromyalgia symptoms, while six thought they had experienced no change, and five believed the symptoms had worsened over the time of the trial. The results of this pilot study suggest that dietary Chlorella supplementation may help relieve the symptoms of fibromyalgia in some patients and that a larger, more comprehensive double-blind, placebo-controlled clinical trial in these patients is warranted.


Subject(s)
Chlorella , Dietary Supplements , Fibromyalgia/therapy , Adult , Dietary Supplements/adverse effects , Female , Fibromyalgia/physiopathology , Humans , Male , Middle Aged , Pilot Projects , Tablets , Treatment Outcome
5.
J Neurooncol ; 45(3): 209-18, 1999.
Article in English | MEDLINE | ID: mdl-10845391

ABSTRACT

We transduced a highly tumorigenic T9 clone (T9.F), isolated from the rat T9 glioblastoma cell line, with a retroviral expression vector containing the human IL-6 cDNA and investigated the effects of IL-6 secretion on glioma formation in the syngeneic Fischer rat. Two subclones producing high and low levels (35 and 3.5 ng/10(6) cells/48 h) of IL-6 were identified and were termed T9.F/IL6/hi and T9.F/IL6/lo, respectively. Subcutaneous (SC) injection of 1 x 10(6) parental T9.F cells resulted in 100% tumor formation and progression. When 1 x 10(6) IL-6 secreting T9.F cells were injected SC, a small palpable tumor formed which sometimes regressed. In this regard, no tumors were detected after 30 days in 76% (13/17) of animals injected with T9.F/IL6/hi cells, whereas only 10% (1/10) of the rats injected with T9.F/IL6/lo cells completely rejected their tumors within this time frame. The addition of an IL-6 neutralizing antibody to the T9.F/IL6/hi SC inoculum followed by an intratumoral injection of the IL-6 neutralizing antibody, seven days later, abrogated the anti-tumor effects. Animals that rejected the IL-6 secreting tumors were 100% protected from subsequent intracranial (IC) challenges with the parental T9.F glioma as well as the original T9 glioblastoma; partially protected from an IC challenge with the unrelated, syngeneic RT-2 glioma; but were not protected from an IC challenge with the syngeneic MadB106 adenocarcinoma. When 1 x 10(4) cells were injected in the brain of naive animals, survival time was significantly increased for those rats implanted with T9.F/IL6/hi cells, but not T9.F/IL6/lo cells, as compared to animals implanted with T9.F parental cells (p = 0.003). This study demonstrates that IL-6 secretion attenuates SC and IC glioma growth and SC rejection of IL-6 secreting T9.F cells induces long-term glioma immunity which is effective in the brain.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Brain Neoplasms/pathology , Carcinogenicity Tests , DNA, Complementary , Female , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Glioblastoma/pathology , Humans , Neoplasm Transplantation , Neutralization Tests , Rats , Rats, Inbred F344 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Retroviridae/genetics , Transduction, Genetic , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/transplantation
6.
Ann N Y Acad Sci ; 890: 42-50, 1999.
Article in English | MEDLINE | ID: mdl-10668412

ABSTRACT

CP-101,606 is a postsynaptic antagonist of the glutamate-mediated NR2B subunit of the N-methyl-D-aspartate (NMDA) receptor. When administered intravenously (i.v.) at the time of injury, CP-101,606 is neuroprotective in animal models of traumatic brain injury (TBI) and ischemia. Minimal adverse effects have been observed in normal human volunteers given i.v. doses of up to 3 mg/kg/hr for 72 hours. The objective of the present clinical trial was to assess the safety, pharmacokinetics, and tolerability of CP-101,606 infused for various times in patients who had suffered either an acute moderate or mild TBI (Glasgow Coma Score 9-14) or hemorrhagic stroke. Patients began receiving treatment within 12 hours of brain injury. A total of 53 subjects (45 with TBI and 8 with stroke) were randomized in a double-blind fashion to receive CP-101,606 or placebo (4 drug: 1 placebo). Drug/placebo was administered by i.v. infusion (0.75 mg/kg/hr) for 2 hours and then stopped (n = 25) or continued for 22 hours (n = 4) or 70 hours (n = 24) at a rate of 0.37 mg/kg/hr. Mean plasma drug concentrations were well above the predicted therapeutic concentration of 200 ng/ml within two hours of initiating treatment and were sustained as long as drug was infused. All the patients tolerated their drug/placebo treatment, and there were no clinically significant cardiovascular or hematological abnormalities in either group. A Neurobehavioral Rating Scale, used to detect personality changes and behavioral disturbances, indicated that all subjects showed an improvement from their postinjury, predosing baseline but did not significantly differ from each other with respect to type of head injury and/or treatment with drug or placebo. Modified Kurtzke Scoring also showed a similar pattern of improvement irrespective of type of head injury or drug/placebo treatment. This study suggests that CP-101,606, infused for up to 72 hours has no psychotropic effects and is well-tolerated in patients who have sustained a mild or moderate TBI or hemorrhagic stroke.


Subject(s)
Brain Injuries/drug therapy , Excitatory Amino Acid Antagonists/administration & dosage , Piperidines/administration & dosage , Stroke/drug therapy , Adolescent , Adult , Aged , Brain Injuries/blood , Double-Blind Method , Excitatory Amino Acid Antagonists/blood , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Piperidines/blood , Stroke/blood
7.
Ann N Y Acad Sci ; 890: 51-8, 1999.
Article in English | MEDLINE | ID: mdl-10668413

ABSTRACT

CP-101,606 is a postsynaptic antagonist of N-methyl-D-aspartate (NMDA) receptors bearing the NR2B subunit. When administered intravenously (i.v.), it decreases the effects of traumatic brain injury (TBI) and focal ischemia in animal models. Therapeutic plasma concentrations (200 ng/ml) in animals, have been well tolerated in healthy human volunteers. The purpose of the present dose escalation study was to assess the safety, tolerability, and pharmacokinetics of CP-101,606 in subjects who had suffered either an acute severe TBI (Glasgow Coma Scale 3-8) or spontaneous intracerebral hemorrhage. Thirty patients, 20 with a TBI and 10 with a stroke, were enrolled in the trial and began receiving an i.v. infusion of CP-101,606 for 2 hours, 24 hours, or 72 hours within 12 hours of brain injury. For the first two hours, the drug was given a rate of 0.75 mg/kg/hr and then stopped (n = 17) or continued for 22 (n = 2) or 70 hours (n = 11) at 0.37 mg/kg/hr. Plasma and cerebrospinal fluid (CSF) were collected at serial times during and after treatment. There were no consistent changes in blood pressure or pulse nor any clinically significant hematological or electrocardiogram (ECG) abnormalities attributable to CP-101,606. No adverse events or behavioral changes were considered to be related to the drug. Plasma concentrations of CP-101,606 over 200 ng/ml were rapidly achieved in the blood and CSF within two hours and were sustained there as long as the drug was infused. CSF concentrations were slightly higher than that in plasma by the end of infusion suggesting good penetration of CP-101,606 into the CSF. Outcome in the severe TBI patients, as measured by the Glasgow Outcome Score at six months, suggested that a two-hour infusion yielded a range of scores similar to contemporary patients with a severe TBI treated at our hospital while the outcomes of the patients treated with either a 24- or 72-hour infusion were better on average. Thus, these results indicate that CP-101,606 infused for up to 72 hours is well tolerated, penetrates the CSF and brain, and may improve outcome in the brain-injured patient.


Subject(s)
Brain Injuries/drug therapy , Cerebral Hemorrhage/drug therapy , Excitatory Amino Acid Antagonists/administration & dosage , Piperidines/administration & dosage , Adolescent , Adult , Aged , Animals , Brain Injuries/blood , Cerebral Hemorrhage/blood , Excitatory Amino Acid Antagonists/blood , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Piperidines/blood , Treatment Outcome
8.
Am J Pathol ; 150(6): 2143-52, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9176405

ABSTRACT

Both melanocytes and glial cells are derived embryologically from the neural ectoderm. Their malignant transformed counterparts, melanoma and glioma cells, respectively, may share common antigens. Numerous tumor-associated antigens have been identified in melanomas but only a few a gliomas. Using an established reverse transcriptase polymerase chain reaction plus Southern blot assay, we compared the mRNA expression of melanoma-associated antigens (MAAs) of melanomas to brain tumors primarily derived from glial cells. The MAAs studied included tyrosinase (Tyr), tyrosinase-related protein-1 and -2 (TRP-1 and TRP-2), gp100, human melanoma antigen-encoding genes 1 and 3 (MAGE-1 and MAGE-3), and melanotransferrin (p97). Glioblastoma multiforme (n = 21), anaplastic astrocytoma (n = 3), ependymoma (n = 2), meningioma (n = 3), oligodendroglioma (n = 1), and melanoma (n = 12) tumor specimens were assayed for MAA mRNA expression. Glioblastoma multiforme, astrocytoma, and melanoma cell lines were also assayed. We observed that individual MAA mRNAs were expressed in these brain tumors and cell lines at varying frequencies. The melanogenesis-pathway-related MAAs Tyr, TRP-1, TRP-2, and gp100 mRNAs were also expressed at different levels in normal brain tissues but at a much lower frequency than in glioblastoma multiforme and melanoma. MAGE-1 and MAGE-3 mRNA were expressed in different types of tumor specimens and cell lines but never in normal brain tissue. Tumor antigen p97 was expressed in all types of tumors and also in normal brain tissues. These studies demonstrate that melanomas and primary brain tumors express common MAAs and could be exploited in patients with malignant glioma by active specific immunotherapy against these common MAAs.


Subject(s)
Antigens, Neoplasm/analysis , Brain Neoplasms/immunology , Glioma/immunology , Intramolecular Oxidoreductases , Melanoma/immunology , Oxidoreductases , Skin Neoplasms/immunology , Blotting, Southern , Brain/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Humans , Isomerases/metabolism , Melanoma/metabolism , Melanoma-Specific Antigens , Membrane Glycoproteins/metabolism , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/metabolism , Proteins/metabolism , Skin Neoplasms/metabolism , Tumor Cells, Cultured , gp100 Melanoma Antigen
9.
Neurol Res ; 19(2): 145-52, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9175143

ABSTRACT

Malignant glioma remains one disease for which there is no curative therapy. Clearly there is a need to explore new and innovative approaches for their treatment. In this report, we review our preclinical trial of a new adoptive immunotherapy protocol using cytotoxic T lymphocytes (CTL) which had been sensitized to glioma in vivo and then activated and their number expanded ex vivo using compounds which enhance signal transduction. These glioma-sensitized lymphocytes, when introduced systemically into rats with either an intracerebral or intradermal glioma, eradicated or slowed the progression of their tumor. These results indicate for the first time that a reproducible and sustained eradication of a malignant glioma could be achieved by the adoptive transfer of tumor-sensitized, ex vivo expanded CTL. A Phase I clinical trial is now underway to test the safety and potential efficacy of this immunotherapy in patients with recurrent malignant glioma.


Subject(s)
Glioma/immunology , Glioma/therapy , Immunization , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , Animals , Brain Neoplasms/therapy , Clinical Trials as Topic , Glioma/pathology , Humans , Lymphocyte Activation , Skin Neoplasms/therapy
10.
J Neurooncol ; 32(1): 19-28, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9049859

ABSTRACT

It has been shown that adoptive immunotherapy can be curative for established malignant tumors. The key to this treatment lies in obtaining sufficient numbers of lymphocytes which are sensitized to recognize tumor antigens and carry out immunological reactions to destroy tumor cells. Reported here are the results of experiments to: 1) sensitize lymphocytes to the antigens of rat glioma cells and expand them ex vivo for use in adoptive immunotherapy, 2) characterize the cells of the expanded population, and 3) evaluate antitumor activity in a cohort of rats with well-established intracranial gliomas. Viable RT-2 glioma cells were injected into the hind foot pads of syngeneic Fischer 344 rats. After 10 days, the tumor draining lymph nodes (DLN) were harvested from the injected limbs and mechanically dissociated. The cells of the DLN were then suspended in culture medium supplemented with low dose interleukin-2 (IL-2) and incubated for 18 hours with Bryostatin-1 and ionomycin (Bryo/Io) to stimulate expansion. The cells were next washed to remove the Bryo/Io and resuspended in culture medium and IL-2. Population expansions of 40- to 100-fold were seen after 8 days. Flow cytometric analysis showed these cells to be a nearly pure population of T lymphocytes of the CD3+CD8+ phenotype. Intravenous injection of the ex vivo expanded DLN cells did not significantly improve survival of rats with a seven-day intracerebral RT-2 glioma, although, compared to untreated controls, the tumors of the treated animals were smaller, showed no necrosis, and appeared to be less infiltrative. Furthermore, the treated animals had a pronounced lymphocytic infiltration of their tumors with greater associated degrees of hemorrhagic change and peritumoral edema. When the ex vivo expanded DLN cells were intravenously injected into three-day intracerebral RT-2 glioma models, tumors were almost always eliminated and the animals survived their tumor challenge. We conclude that successful expansion of glioma-sensitized DLN lymphocytes is possible and that adoptive immunotherapy using these cells is capable of effectively limiting the progression of large gliomas, while totally eradicating small ones.


Subject(s)
Brain Neoplasms/immunology , Cell Communication/drug effects , Glioma/immunology , Lymph Nodes/ultrastructure , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Animals , Antigens, Neoplasm/immunology , Brain Neoplasms/ultrastructure , Female , Glioma/ultrastructure , Immunotherapy, Adoptive , Interleukin-2/pharmacology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Phenotype , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
11.
J Neurooncol ; 32(1): 29-38, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9049860

ABSTRACT

We have investigated the anti-tumor activity of ex vivo activated and expanded T cells which had been sensitized in vivo to one of two different syngeneic rat glioma cell lines; D74 or RT-2. Rats were sensitized by inoculation of irradiated tumor cells into each hind foot pad. After 10 days, the tumor-draining lymph node (DLN) from each popliteal region was excised and prepared as a single cell suspension. Tumor-DLN lymphocytes were next activated overnight in RPMI-1640 medium containing 10% fetal bovine serum (FBS), Bryostatin-1 (5 nM), ionomycin (1 microM), and 20 U human recombinant interleukin-2 (IL-2) per ml. Culture for seven days in RPMI-1640 supplemented with FBS and IL-2 resulted in approximately 100-fold expansion of the lymphocyte population. Both D74- and RT-2-sensitized T cells constitutively secreted tumor necrosis factor-alpha, and both lymphocyte populations produced comparable amounts of the cytokine when co-cultured with either glioma cell line. Neither D74- and RT-2-sensitized effectors constitutively secreted gamma-interferon (gamma-IFN), but both populations produced gamma-IFN when exposed to either glioma cell line in vitro. D74-sensitized T cells released significantly more gamma-IFN than the RT-2 DLN lymphocytes. In vitro Chromium-release assays indicated that RT-2-sensitized T cells were more cytotoxic for RT-2 targets than for the D74 line and that D74-sensitized effectors were also more cytotoxic for RT-2 targets. To assess in vivo therapeutic efficacy, rats who had been inoculated intradermally with RT-2 cells three days earlier received an intravenous injection of RT-2- or D74-sensitized DLN cells (10(6) cells/gram body weight) expanded after activation with Bryostatin-1 and ionomycin or an equal number of lymphokine-activated killer (LAK) cells. Tumor diameters were measured daily and revealed that injection of glioma-sensitized lymphocytes led to the elimination of tumor while treatment with LAK cells had no therapeutic benefit. These results indicate, that at least for these two glioma lines, gamma-IFN release, rather than in vitro cytotoxicity, was a better predictor for in vivo immunotherapeutic efficacy of the glioma-sensitized, expanded T cells.


Subject(s)
Brain Neoplasms/immunology , Cell Communication/drug effects , Cytokines/biosynthesis , Glioma/immunology , Lymph Nodes/ultrastructure , Signal Transduction/drug effects , T-Lymphocytes/metabolism , Animals , Brain Neoplasms/ultrastructure , Chromium Radioisotopes , Female , Glioma/ultrastructure , Immunotherapy, Adoptive , Interferon-gamma/biosynthesis , Lymph Nodes/cytology , Lymph Nodes/drug effects , Rats , Rats, Inbred F344 , T-Lymphocytes/immunology , Tumor Cells, Cultured
12.
Acta Neuropathol ; 93(1): 78-86, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9006660

ABSTRACT

The pro-inflammatory and blood-brain barrier (BBB) effects of intratumoral (IT) injection of human recombinant tumor necrosis factor-alpha (rTNF-alpha) were studied in the Fischer rat RT-2 glioma model. Animals received a single stereotaxic injection of either 6 x 10(4) U rTNF-alpha or excipient (vehicle) into the center of an intracerebrally implanted glioma. In order to demonstrate any effects rTNF-alpha might have on the BBB, studies were conducted using endogenous IgG (150 kD) as a tracer. Forty-eight hours following injection of excipient, a margin of peritumoral IgG extravasation was observed while rats treated with 6 x 10(4) U rTNF-alpha showed a dense and extensive IgG extravasation involving both hemispheres. Histological examination revealed that an IT rTNF-alpha injection induced leukocytic adherence, neutrophilic cuffing and infiltration throughout the lesion from 12 to 72 h after injection. These histological observations were supported by quantification of cerebral myeloperoxidase (MPO) levels which indicated a significant increase in neutrophils over the excipient recipients at 4 and 12 h. These MPO levels contrasted with our earlier studies in normal rats which revealed no significant difference in tissue MPO levels following injection of excipient or rTNF-alpha. In addition, when MPO levels in tumor models and normal rats receiving TNF were compared, a significantly greater presence of neutrophils was seen in tumor models at 12 h post-TNF injection. We believe that the increased inflammatory response seen in a progressing glioma compared to normal brain may be the result of decreased resistance to leukocytic infiltration due to increased vascular surface area, the lack of infiltration-resistant perivascular basement membrane, and/or increased extracellular space.


Subject(s)
Brain Neoplasms/therapy , Capillary Permeability/drug effects , Cell Movement/drug effects , Cerebrovascular Circulation/drug effects , Glioma/therapy , Leukocytes/drug effects , Recombinant Proteins/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Adhesion , Cerebral Hemorrhage/pathology , Endothelium, Vascular/pathology , Glioma/enzymology , Glioma/pathology , Humans , Immunoglobulin G/metabolism , Injections, Intralesional , Injections, Intraventricular , Leukocytes/pathology , Peroxidase/metabolism , Rats , Rats, Inbred F344 , Recombinant Proteins/pharmacokinetics , Tumor Necrosis Factor-alpha/pharmacokinetics
13.
Toxicology ; 95(1-3): 177-85, 1995 Jan 06.
Article in English | MEDLINE | ID: mdl-7825184

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is one of the most potent environmental immunomodulating agents identified so far. Historically, mice have been used to model mammalian immunobiology and most of the data gathered on the immunotoxicity of TCDD has been obtained from studies with mice. However, rats have been used more extensively in toxicological research to establish human risk assessment criteria. A need exists, therefore, to develop a database using the rat model in immunotoxicology so that complete animal toxicity studies can be conducted. We have treated female Fischer 344 rats with a single i.p. dose of 0.3, 3.0, or 30.0 micrograms/kg TCDD or corn oil vehicle and examined cytotoxic T-cell (CTL) activities 24 days following treatment. Syngeneic in vivo tumor-specific CTLs were generated that model cell-mediated immune reactions against neoplastically transformed self antigens. RT2, a virally-induced Fischer 344 rat glioma, and D74, a ethylnitrosurea-induced Fischer 344 rat glioma were used as targets. This immunological parameter was compared to body, thymic, and liver weights as well as liver ethoxyresorufin deethylase (EROD) activity on day 24 post-TCDD treatment. The results indicate that Fischer 344 rats are very sensitive to TCDD as indicated by severe thymic atrophy and EROD induction at all three doses. In contrast, CTL activity was only marginally affected by these same doses of TCDD with only a modest suppression noted at the highest dose. These results indicate that the CTL response in rats may not be useful in characterizing the effects of this xenobiotic on immunocompetence in the rat.


Subject(s)
Polychlorinated Dibenzodioxins/toxicity , T-Lymphocytes, Cytotoxic/drug effects , Animals , Body Weight/drug effects , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic/drug effects , Female , Liver/drug effects , Rats , Rats, Inbred F344 , T-Lymphocytes, Cytotoxic/immunology
14.
J Neurooncol ; 20(1): 17-25, 1994.
Article in English | MEDLINE | ID: mdl-7807180

ABSTRACT

Human recombinant tumor necrosis factor-alpha (rTNF-alpha) was administered to normal Fischer 344 rats by stereotaxic intracerebral (IC) injection. Animals receiving a single injection of either 6 x 10(4) U rTNF-alpha or an equivalent volume of excipient (vehicle) in their right parietal lobe. In order to demonstrate any effects rTNF-alpha might have on the blood-brain barrier (BBB), two studies were conducted, one employing exogenous horseradish peroxidase (HRP,44 kD) as a tracer of BBB permeability and the other using endogenous IgG (150 kD). Rats given rTNF-alpha showed transitory BBB permeability to HRP by 24 hours post-injection; this BBB compromise was determined to be no longer than 60 hours. In the other study, IgG was seen to cross the BBB by 48 hours post-rTNF-alpha injection. Alternatively, rats injected IC with excipient showed only limited BBB opening as a result of injection-induced trauma. We conclude that human rTNF-alpha, injected IC into normal rats triggers a temporary breakdown in BBB integrity which begins sometimes between 12 and 24 hours post-injection, is large enough to permit macromolecules of at least 150 kD to pass, and resolves by 72 hours post-injection.


Subject(s)
Blood-Brain Barrier/drug effects , Animals , Female , Horseradish Peroxidase/analysis , Humans , Immunoglobulin G/analysis , Rats , Rats, Inbred F344 , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology
15.
Neurosurgery ; 34(1): 129-35, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8121549

ABSTRACT

Nervous system-specific transcription factors that bind to the octameric deoxyribonucleic acid sequence motif ATGCAAAT (or ATTTGCAT) are known as N-Oct proteins. Neurons and glia contain the ubiquitous Oct-1 protein and four polypeptide complexes termed N-Oct-2, N-Oct-3, N-Oct-4, and N-Oct-5. Previously, we showed that N-Oct proteins are differentially expressed by human neuroblastoma and glioblastoma cell lines in vitro. We have now extended this work to freshly isolated human primary and metastatic brain tumors. Contrary to brain tumor cell lines, of the five astrocytomas and three glioblastomas analyzed, all but two tumors displayed the complete N-Oct protein profile, irrespective of histopathological tumor grade. Two astrocytomas were negative for N-Oct-4. Ten of 13 ependymomas exhibited N-Oct-2, N-Oct-3, and N-Oct-4 but lacked the N-Oct-5 complex. In contrast, brain metastases of two patients with extracerebral carcinomas contained only Oct-1, and cerebral metastases from two cases of B cell lymphomas showed Oct-1 and Oct-2 complexes, the characteristic Oct protein pattern of B lymphocytes. Thus, metastatic carcinoma and lymphoma expressed a non-nervous system phenotype of Oct proteins.


Subject(s)
Brain Neoplasms/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins , Transcription Factors/genetics , Astrocytoma/genetics , Astrocytoma/pathology , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Ependymoma/genetics , Ependymoma/pathology , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Glioblastoma/pathology , Homeodomain Proteins , Humans , Neoplasm Staging , Octamer Transcription Factor-2 , Octamer Transcription Factor-3 , Oligodendroglioma/genetics , Oligodendroglioma/pathology , POU Domain Factors
16.
Neurosurgery ; 31(1): 89-98; discussion 98-9, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1641114

ABSTRACT

It is well documented that drug delivery into experimental and human brain tumors is limited by the variably intact blood-brain barrier (BBB) at the growing edge. The aim of the present investigation was to examine the histopathological changes that occur after a single intralesional injection of human recombinant interleukin-2 (rIL-2) into a growing glioma and determine whether the injection improved delivery of cytotoxic drug into the neuropil surrounding the site of lymphokine injection. Because an intracerebral injection of rIL-2 causes a temporary breakdown in the BBB, we hoped to enhance drug penetration into peritumoral areas of brain with an intact BBB by using the novel biomodulating effect of rIL-2 on the cerebral endothelial cells. The results demonstrated that an intralesional injection of 7.2 x 10(4) National Units rIL-2 on Day 7 after tumor inoculation did not accentuate the already increased cerebrovascular permeability produced by the glioma nor did rIL-2 trigger additional or aggravate neurological deficits in glioma-bearing rats. Before the administration of chemotherapy in vivo, the RT-2 glioma cells were tested for in vitro sensitivity by colorimetric assay. At 24 hours after exposure to either methotrexate (MTX), vincristine (VIN), or doxorubicin (DOX), no significant inhibition of metabolic activity was observed. In contrast, a timed pulsed of any drug for 5 minutes caused significant dose-dependent inhibition of RT-2 glioma cells at 48 hours to 5 days after drug administration. Animal models receiving an intralesional injection of rIL-2 followed 3 days later by an intravenous dose of 30 mg/kg MTX, 0.23 mg/kg VIN, or 10 mg/kg DOX demonstrated that only MTX combined with intralesional rIL-2 significantly inhibited intracranial proliferation of RT-2 glioma cells. Use of intralesional rIL-2 and intravenous chemotherapy, however, did not significantly increase survival in this animal model of glioma. These results show that the combination of cytotoxic drugs with intralesional rIL-2 can be safely applied in the management of glioma and may form a rational basis for additional pharmacological investigations of a wider assortment of chemotherapies in combination with rIL-2 for intracranial malignancies.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/therapy , Brain/blood supply , Cell Division/drug effects , Doxorubicin/administration & dosage , Glioma/therapy , Interleukin-2/administration & dosage , Methotrexate/administration & dosage , Vincristine/administration & dosage , Animals , Blood-Brain Barrier/physiology , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Cell Division/physiology , Cell Line , Cell Survival/drug effects , Combined Modality Therapy , Doxorubicin/pharmacokinetics , Glioma/pathology , Glioma/physiopathology , Humans , Injections, Intralesional , Interleukin-2/pharmacokinetics , Methotrexate/pharmacokinetics , Necrosis , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Vincristine/pharmacokinetics
17.
J Neurooncol ; 13(1): 43-55, 1992 May.
Article in English | MEDLINE | ID: mdl-1613537

ABSTRACT

We investigated the effects of daily subcutaneous (SC) injections of 100, 200, or 400 micrograms/kg murine recombinant interleukin-1 beta (rIL-1 beta) or its excipient on normal Fischer 344 rats and ones harboring a malignant RT-2 glioma. The tumor model has a predictable course with animals dying on days 14-17 following an intracerebral inoculation of 10(4) RT-2 glioma cells. Treatments with rIL-1 beta or excipient began on day seven post-tumor inoculation and continued for 7 days. We observed no significant effect on core body temperatures although there was a significant (p less than 0.05) decrease in body weight in all rIL-1 beta treated animals. When tumor-bearing animals became moribund, they received an intraperitoneal injection of bromodeoxyuridine (BUdr) and were sacrificed two hours later. Blood samples were obtained prior to their sacrifice by transcardiac perfusion with a buffered aldehyde solution. Recombinant IL-1 beta affected blood differentials; causing neutrophilia, lymphopenia, and slight thrombocythemia. The BUdr labeling index of glioma cells did not significantly differ between treatment groups, although tumors differed histologically at the time of necropsy. Tumors of rIL-1 beta treated animals had more extensive necrosis and a greater degree of leukocyte infiltration. Survival studies were conducted in which rats were given continuous daily SC injections of rIL-1 beta until day of death. Overall survival between the two groups differed significantly in studies using 100 micrograms/kg/d (p less than 0.05); rIL-1 beta treated rats had a mean survival time of 22 (+/- 3.0) days while excipient controls had a mean survival time of 17 (+/- 0.5) days. Similarly, at a dose of 200 micrograms rIL-1 beta/kg/d, mean survival was significantly (p less than 0.05) increased as compared to excipient controls (18.75 +/- 1.5 vs. 15.25 +/- 1.7 days, respectively). Daily injections of 400 micrograms/kg did not significantly increase the survival of glioma bearing animals, possibly as a consequence of rIL-1 beta toxicity at this dose.


Subject(s)
Glioma/drug therapy , Interleukin-1/therapeutic use , Animals , Bromodeoxyuridine , Cell Division/drug effects , Cell Line , Female , Glioma/blood supply , Glioma/pathology , Interleukin-1/toxicity , Mice , Rats , Rats, Inbred F344 , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity
18.
Cell Immunol ; 140(1): 145-57, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1739983

ABSTRACT

Although there is evidence that corticosteroids inhibit receptor-ligand-induced phospholipid hydrolysis, the immunosuppressive effects of these agents downstream of protein kinase C (PK-C) activation and cytosolic Ca2+ mobilization is unclear. Previous studies indicated that T cell proliferative activation could be achieved with simultaneous short-term (e.g., 15-120 min) exposure to agents activating PK-C and elevating cytosolic Ca2+. In the studies reported here, similar procedures were utilized for determining whether corticosteroids alter T cell activation signals downstream of second messenger events. Dexamethasone interfered with T cell activation induced by short-term exposure to phorbol 12,13-dibutyrate (PDBu) and the calcium ionophore, ionomycin. The inhibitory effect was evident with as little as 15 min of exposure to dexamethasone and T cell activating agents, making mechanisms involving de novo protein synthesis unlikely. Dexamethasone's effects in this system were blocked by the steroid receptor antagonist RU-486, indicating that the inhibition was mediated through the glucocorticoid receptor. The inclusion of recombinant interleukin-2 (IL-2) only partially overcame the dexamethasone inhibitory effect. Long-term (i.e., 48 hr) direct stimulation of PK-C with either PDBu or the non-tumor-promoting PK-C activator, bryostatin 1, also substantially overcame dexamethasone's effects, resulting in a recovery of IL-2 production and significant restoration of the T-cell proliferative response. These observations suggest that treatment with a PK-C-activating agent such as bryostatin 1 could reduce glucocorticosteroid-induced immunosuppression.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Ionomycin/pharmacology , Lymphocyte Activation/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , T-Lymphocytes/drug effects , Bryostatins , Calcium/metabolism , Cell Division/drug effects , Dexamethasone/antagonists & inhibitors , Dexamethasone/pharmacology , Drug Interactions , Enzyme Activation/drug effects , Humans , Interleukin-2/analysis , Lactones/pharmacology , Macrolides , Mifepristone/pharmacology , Monocytes/drug effects , Protein Kinase C/metabolism , Second Messenger Systems , Time Factors
19.
J Neurosurg ; 76(2): 251-60, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1730954

ABSTRACT

Patients harboring a malignant brain tumor have been described as being highly immunosuppressed, as evidenced by reduced numbers of T cells and the decreased ability of their lymphocytes to produce interleukin-2 (IL-2). In order to determine whether an intrinsic abnormality exists in the T lymphocytes of glioma patients and to evaluate what role corticosteroids may play in glioma-associated immunosuppression, in vitro T cell proliferative function in the presence of recombinant IL-2 (rIL-2) was examined in age-matched groups of normal control subjects, steroid-free patients with glial tumors, steroid-dependent patients with glial tumors, and steroid-dependent patients with nonglial cerebral tumors. The results demonstrated that, when enriched T cell populations of all brain-tumor patients were stimulated with rIL-2 and phytohemagglutinin (PHA), there were no statistically significant differences between any groups. In contrast, when T cell populations were stimulated with mitogenic combinations of phorbol ester, calcium ionophore, and rIL-2, those from steroid-dependent patients with glial tumors had a significantly lower response than those from normal control subjects, suggesting that a population of T cells capable of responding to phorbol ester/ionomycin and not PHA stimulation is inhibited by corticosteroid therapy in glioma patients. In addition, T cells of four brain-tumor patient/age-matched control subject pairs were stimulated with either phorbol ester/ionomycin or PHA for 24 hours; three of the four patients expressed low-affinity IL-2 receptor levels as high or higher than their respective control subjects, suggesting that IL-2 receptor expression in these patients may be quantitatively normal once the T cell number is corrected. Taken together, these results show that the decreased PHA responsiveness that has been previously reported in lymphocytes of glioma patients is not due to a cellular abnormality within the potentially responsive cells, but rather reflects the reduced proportion of T cells within their peripheral blood which, as a consequence, reduces the level of IL-2 production attained upon activation.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Female , Humans , Immune Tolerance/drug effects , Interleukin-2/pharmacology , Ionomycin/pharmacology , Lymphocyte Activation/drug effects , Male , Middle Aged , Phorbol 12,13-Dibutyrate/pharmacology , Phytohemagglutinins/immunology , Receptors, Interleukin-2/immunology , Recombinant Proteins , T-Lymphocytes/drug effects , Tumor Cells, Cultured
20.
J Neurooncol ; 12(1): 75-83, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1541981

ABSTRACT

Nine patients with a recurrent malignant glioma were treated with repeated intracavitary or intracerebroventricular injections of human recombinant interleukin-2 (rIL-2) alone or in combination with systemic interferon-alpha (IFN-alpha). Five patients received only rIL-2 and four were treated with rIL-2 plus subcutaneous injections of IFN-alpha. Therapy was administered on a Monday, Wednesday, Friday schedule for up to 10 weeks, beginning with a dose of 10,000 IU rIL-2/injection. Doses were escalated every two weeks until some toxicity was apparent. The maximum amount of rIL-2 any one patient in this group received was 580,000 IU. Patients on combination immunotherapy were held at an rIL-2 dosage of 10,000 IU while IFN-alpha, which began at 3 million IU, was escalated every other week up to 18 million IU/dose. They were then held at that IFN-alpha dosage and rIL-2 was increased to 50,000 IU. The total amount of rIL-2 and IFN-alpha any one in this group received was 510,000 IU and 417 million IU, respectively. Repeated injections of 10,000 IU rIL-2 were well-tolerated by all nine patients and no change in their functional status was seen. At doses at 50,000 IU rIL-2, increased edema around the tumor cavity was observed by MRI/CT scand in 3/5 patients and clinical side-effects in the form of somnolence and headache along with some morbidity specifically associated with tumor location were also seen. Patients receiving rIL-2+ IFN-alpha showed progressive fatigue, muscle weakness, and occasionally nausea. Two of these patients showed increased peritumoral edema on MRI/CT scan.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Interferon Type I/therapeutic use , Interleukin-2/therapeutic use , Adult , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Drug Administration Schedule , Drug Evaluation , Female , Glioma/diagnostic imaging , Glioma/surgery , Humans , Injections , Injections, Intraventricular , Interferon Type I/administration & dosage , Interferon Type I/toxicity , Interleukin-2/administration & dosage , Interleukin-2/toxicity , Male , Neoplasm Recurrence, Local , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...