Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Biotechniques ; 30(1): 202-6, 208, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11196312

ABSTRACT

Microarray technology is a powerful tool for generating expression data on a large number of genes simultaneously. However, as for any assay, it must be reproducible to give confidence in the results. Using a classical statistical method--the factorial design of experiments--we have assessed the effects of different experimental factors in our system. Significant effects on signal were seen when the standard components were substituted with a different enzyme, fluorescent label, or RNA purification method. This has led to the implementation of an improved procedure that maximizes signal without affecting the variability of the system, thus increasing the signal-to-noise ratio. In addition, we were able to quantify the variability between microarrays and replicates within microarrays.


Subject(s)
DNA, Complementary/genetics , Oligonucleotide Array Sequence Analysis/methods , Analysis of Variance , Animals , DNA Probes , Liver/metabolism , Nucleic Acid Hybridization , RNA/genetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results
2.
Clin Cancer Res ; 6(10): 4148-53, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11051269

ABSTRACT

Intracerebral microinfusion (ICM) is an innovative technique of delivering therapeutic agents throughout large portions of the brain that circumvents the blood-brain barrier, minimizes systemic toxicity, and provides a homogeneous distribution of the infused agent. Temozolomide is a novel methylating agent with proven efficacy against malignant gliomas (MGs) after systemic administration but with dose-limiting myelotoxicity. Because MGs rarely metastasize, systemic drug delivery is unnecessary. Therefore, we evaluated the efficacy and toxicity of ICM with temozolomide in an athymic rat model of human MGs. Treatment of rats by ICM with temozolomide 3 days after intracerebral challenge with D54 human MG xenograft increased median survival by 128% compared with rats treated by ICM with saline, by 113% compared with rats treated with i.p. saline, and by 100% compared with rats treated with i.p. temozolomide (P < 0.001). Delay of treatment until 9 days after tumor challenge still resulted in a 23% increase in median survival in rats treated by ICM of temozolomide compared with rats treated with i.p. temozolomide. In addition, overall, 21.7% of rats treated by ICM with temozolomide survived for > 100 days without clinical or histological evidence of tumor. The dose of temozolomide delivered by ICM in this study was limited only by drug solubility, and no neurological or systemic toxicity could be attributed to ICM with temozolomide. Therefore, ICM of temozolomide may offer significant advantages in the treatment of MGs.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Catheters, Indwelling , Dacarbazine/analogs & derivatives , Dacarbazine/administration & dosage , Drug Delivery Systems , Glioma/drug therapy , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Brain/drug effects , Catheterization , Dacarbazine/therapeutic use , Humans , Male , Neoplasm Transplantation , Rats , Rats, Nude , Temozolomide , Time Factors , Tumor Cells, Cultured
3.
Proc Natl Acad Sci U S A ; 97(13): 7503-8, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10852962

ABSTRACT

The epidermal growth factor receptor (EGFR) is often amplified and rearranged structurally in tumors of the brain, breast, lung, and ovary. The most common mutation, EGFRvIII, is characterized by an in-frame deletion of 801 base pairs, resulting in the generation of a novel tumor-specific epitope at the fusion junction. A murine homologue of the human EGFRvIII mutation was created, and an IgG2a murine mAb, Y10, was generated that recognizes the human and murine equivalents of this tumor-specific antigen. In vitro, Y10 was found to inhibit DNA synthesis and cellular proliferation and to induce autonomous, complement-mediated, and antibody-dependent cell-mediated cytotoxicity. Systemic treatment with i.p. Y10 of s.c. B16 melanomas transfected to express stably the murine EGFRvIII led to long-term survival in all mice treated (n = 20; P < 0.001). Similar therapy with i.p. Y10 failed to increase median survival of mice with EGFRvIII-expressing B16 melanomas in the brain; however, treatment with a single intratumoral injection of Y10 increased median survival by an average 286%, with 26% long-term survivors (n = 117; P < 0.001). The mechanism of action of Y10 in vivo was shown to be independent of complement, granulocytes, natural killer cells, and T lymphocytes through in vivo complement and cell subset depletions. Treatment with Y10 in Fc receptor knockout mice demonstrated the mechanism of Y10 to be Fc receptor-dependent. These data indicate that an unarmed, tumor-specific mAb may be an effective immunotherapy against human tumors and potentially other pathologic processes in the "immunologically privileged" central nervous system.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , ErbB Receptors/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neoplasm/administration & dosage , Brain Neoplasms/drug therapy , Cytotoxicity, Immunologic , ErbB Receptors/immunology , Female , Humans , Mice , Mutation , T-Lymphocytes/immunology
4.
J Neuroimmunol ; 103(1): 16-25, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10674985

ABSTRACT

To evaluate the efficacy and toxicity of dendritic cell (DC) based therapy for intracerebral gliomas, we utilized a cell line derived from an astrocytoma that arose spontaneously in a VM/Dk mouse. This astrocytoma mirrors human gliomas phenotypically, morphologically and secretes transforming growth factor (TGF)-betas, immunosuppressive cytokines secreted by human gliomas. Systemic vaccination of mice with DCs pulsed with tumor homogenate followed by intracranial tumor challenge produced a > 160% increase in median survival (p = 0.016) compared with mice vaccinated with PBS or unpulsed DCs (p = 0.083). Fifty percent of mice treated with pulsed DCs survived long-term. Immunologic memory was demonstrated by survival of mice rechallenged with tumor. Both cell-mediated and humoral immunity was induced. On histological examination only focal areas of demyelination at the tumor implantation site were present. There was no evidence that autoimmune encephalomyelitis was induced by DC vaccination. Therefore, in a murine model, vaccination with DCs pulsed with glioma tumor homogenate is a safe and effective therapy against a syngeneic glioma located in the immunologically privileged central nervous system (CNS).


Subject(s)
Bone Marrow Cells/immunology , Brain Neoplasms/immunology , Dendritic Cells/immunology , Glioma/immunology , Animals , Brain Neoplasms/therapy , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Glioma/therapy , Mice , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccination
5.
Environ Mol Mutagen ; 34(2-3): 131-9, 1999.
Article in English | MEDLINE | ID: mdl-10529737

ABSTRACT

The mutagenicity and clastogenicity of the immunosuppressive drug azathioprine (AZA), a multitissue rodent carcinogen and IARC-classified human carcinogen, was investigated using transgenic lacZ mice (MutaMouse). Male animals (n = 5 per group) were dosed with AZA (10, 50, 100 mg/kg p.o. daily for 5 days), vehicle (n = 10), or the positive control, chlorambucil (15 mg/kg i.p., n = 3), and killed 24 hr or 25 days after the last treatment. Micronucleus assays were performed with bone marrow (24-hr samples) or peripheral blood (24-hr and 25-day samples) and DNA was extracted from bone marrow and liver for gene mutation analysis at the transgenic lacZ locus. AZA induced 5.3-111.3-fold increases in %MNPCE (P < 0.01) in bone marrow compared with vehicle control, accompanied by 4.4-5. 6-fold increases in %MNRETs (P < 0.01) in peripheral blood. Chlorambucil caused a 14.5-fold increase in %MNRET and there was evidence of significant stem cell toxicity in both positive control and AZA treatment groups. By day 25, however, there was evidence of substantial recovery of the bone marrow as determined by the frequency of RET, and the %MNRET in all treatment groups was the same as the vehicle control. Analysis of lacZ MF showed 1.4-1.6-fold increases in AZA 24-hr bone marrow samples, increasing to approximately 2.0-fold above concurrent controls by day 25 (medium dose P < 0.05, high dose P < 0.01). For liver, there was a 2-fold increase in MF (P < 0.05) in the 24-hr sample at the highest dose only, and increases of 1.3-1.5-fold by day 25 in the medium (P < 0. 05) and high (P = 0.055) dose groups, respectively. The positive control, chlorambucil, induced 2-3-fold increases (P < 0.01) in mean MF in both bone marrow (25-day sample) and liver (24-hr and 25-day samples). These data confirm the clastogenicity of AZA in the mouse, and show that this compound induces gene mutations in bone marrow and liver, in vivo, at the highest dose and supports the view that AZA is a genotoxic carcinogen.


Subject(s)
Azathioprine/toxicity , Mutagens/toxicity , Mutation , Animals , Bone Marrow/drug effects , Bone Marrow/ultrastructure , Liver/drug effects , Male , Mice , Mice, Mutant Strains , Micronucleus Tests
6.
Int J Radiat Oncol Biol Phys ; 45(2): 491-9, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10487576

ABSTRACT

PURPOSE: The antitenascin human/mouse chimeric monoclonal antibody labeled with the alpha-particle-emitting radionuclide 211At is of interest as an endoradiotherapeutic agent for the treatment of brain tumors. To facilitate the investigation of 211At-labeled chimeric 81C6 in patients, the long-term radiotoxicity of this radiopharmaceutical has been evaluated. METHODS AND MATERIALS: Antibody labeling was performed using N-succinimidyl 3-[211At]astato-benzoate. After an initial dose-finding experiment, a second toxicity study was carried out at 4 dose levels in groups of 30 nonthyroid blocked B6C3F1 mice per group (15 males, 15 females). Male mice received either saline or 15-81 kBq/g and females received either saline or 16-83 kBq/g of 211At-labeled antibody. Ten animals (5 males, 5 females) were followed for 6 months and the remainder for 1 year. RESULTS: The lethal dose in 10% of animals (LD10) for 211At-labeled chimeric 81C6 was 46 kBq/g in females and 102 kBq/g in males. Toxic effects--perivascular fibrosis of the intraventricular septum of the heart, bone marrow suppression, splenic white pulp atrophy, and spermatic maturational delay--generally were confined to a few animals receiving the highest doses of labeled antibody. CONCLUSIONS: The LD10 of 211At-labeled chimeric 81C6 in this mouse strain was about half that of [211At]astatide. These results establish the preclinical maximum tolerated dose of 211At-labeled chimeric 81C6 and define in the mouse the target organs for toxicity. These studies will be useful for determining starting doses for clinical studies with 211At-labeled chimeric 81C6.


Subject(s)
Astatine/therapeutic use , Immunoglobulin G/therapeutic use , Immunotoxins/therapeutic use , Radiopharmaceuticals/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Astatine/administration & dosage , Astatine/immunology , Drug Screening Assays, Antitumor , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Immunotoxins/administration & dosage , Male , Mice , Organ Specificity , Radiopharmaceuticals/administration & dosage , Sex Factors
7.
Clin Cancer Res ; 5(9): 2646-52, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499644

ABSTRACT

The incidence of neoplastic meningitis is on the rise. Neoplastic meningitis can result from a direct seeding of the neuraxis by primary brain tumors or by hematogeneous spread of systemic solid tumors. A frequent genetic alteration in primary brain tumors such as gliomas is an in-frame deletion in the epidermal growth factor receptor (EGFR) gene EGFRvIII, which brings together what were normally distant polypeptide sequences in the intact receptor. A novel glycine is formed at the fusion junction, resulting in a unique and tumor-specific target. By using phage display, we have isolated a single-chain antibody specific for the EGFRvIII mutation and expressed it with a modified form of the Pseudomonas exotoxin to form the immunotoxin MR1scFvPE38KDEL (MR-1). The multiple dose toxicity and therapeutic efficacy of MR-1 immunotoxin were tested in an athymic rat model of neoplastic meningitis. The maximally tolerated doses in non-tumor-bearing rats were three doses of 3 microg each. For therapeutic studies, the target was a neoplastic meningitis induced by intrathecal inoculation of the EGFRvIII-expressing human glioma U87MG.deltaEGFR. A dose escalation study compared the survival of three equal doses of 1, 2, and 3 microg of MR-1 immunotoxin with saline or 3 microg of the control immunotoxin specific for the interleukin 2 receptor, anti-Tac. All animals treated with three doses of saline or 3 microg of anti-Tac died, with median survival of 7 and 10 days, respectively. There were 75% (six of eight) long-term survivors in the group treated with three doses of 1 microg and 57% (four of seven) long-term survivors in the groups treated with three doses of either 2 or 3 microg of MR-1 immunotoxin. None of the MR-1 immunotoxin-treated groups reached median survival by the termination of the study at 53 days. Therefore, median survival was estimated to be >53 days, resulting in an estimated increase in median survival of >657% compared with saline and 430% versus anti-Tac. Compartmental therapy with three doses of 2 microg of MR-1 immunotoxin is effective in the treatment of EGFRvIII-expressing neoplastic meningitis. This dose was found to have no clinical or histopathological effects on non-tumor-bearing animals. MR-1 immunotoxin is, therefore, considered specific and safe within its therapeutic window. Phase I clinical trials for tumors invading the intrathecal space that express the EGFRvIII target should be initiated.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , ErbB Receptors/immunology , ErbB Receptors/metabolism , Exotoxins/pharmacology , Immunoglobulin Variable Region/pharmacology , Immunotoxins/pharmacology , Meningeal Neoplasms/drug therapy , Virulence Factors , Animals , Antibody Specificity , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Exotoxins/toxicity , Female , Humans , Immunoglobulin Variable Region/toxicity , Immunotoxins/toxicity , Injections, Spinal , Meningeal Neoplasms/metabolism , Mice , Mice, Nude , Mutation , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Nude , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
8.
Clin Cancer Res ; 5(5): 1183-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10353755

ABSTRACT

Neoplastic meningitis (NM) results from leptomeningeal dissemination of cancers arising within the central nervous system or metastasizing to the leptomeninges from systemic neoplasms. The inability to produce therapeutic drug levels intrathecally (i.t.) with systemic administration and the minimal efficacy of chemotherapeutic agents currently available for direct i.t. use limit therapy. Temozolomide [8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4([3H])-one] is a novel methylating agent with proven activity against intraparenchymal malignant gliomas (MGs). Insolubility of the standard formulation prevents its efficacious use as an i.t. agent, however. To overcome this obstacle, we have developed a unique microcrystalline formulation of temozolomide with greatly enhanced solubility. Treatment of athymic rats bearing subarachnoid MER- human MG xenografts with four doses of i.t. microcrystalline temozolomide over a 2-week period produced a 142% increase in median survival at individual doses of 2.2 micromol (P = 0.0073) and a >367% increase in median survival at individual doses of 6.8 micromol (P = 0.0015). At the higher dose tested, three of eight rats treated developed no neurological symptoms and had no evidence of residual tumor on histological examination after treatment. Use of this microcrystalline formulation in athymic rats bearing subarachnoid MER+ human MG xenografts increased median survival >132% (P < 0.0058) at both dose levels tested. Toxicity directly attributable to the i.t. administration of microcrystalline temozolomide was exhibited in the highest dose groups only and was limited to small patchy areas of focal demyelination involving <5% of spinal cord long tracks.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Meningeal Neoplasms/drug therapy , Meningeal Neoplasms/secondary , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/toxicity , Dacarbazine/administration & dosage , Dacarbazine/pharmacokinetics , Dacarbazine/therapeutic use , Dacarbazine/toxicity , Drug Screening Assays, Antitumor , Humans , Neoplasm Transplantation , Rats , Rats, Nude , Solubility , Subarachnoid Space , Temozolomide , Transplantation, Heterologous
9.
J Neurooncol ; 44(3): 233-41, 1999.
Article in English | MEDLINE | ID: mdl-10720203

ABSTRACT

The current study was designed to evaluate the toxicity and activity of Spartaject Busulfan, a microcrystalline preparation of busulfan, following its intrathecal administration into a nude rat model of human neoplastic meningitis. Animals were treated through permanent indwelling subarachnoid catheters. Human glioma D-456 MG growing in the subarachnoid space was treated with 8.1 micromol of intrathecal Spartaject Busulfan. Single-dose therapy was also subsequently compared with 4 doses of 8.1 and 2.0 micromol busulfan, respectively, against D-456 MG neoplastic meningitis. Additional experiments evaluated a saline control versus 8.1 micromol x 1, 6.2 micromol x 4 and 4.1 micromol x 4, respectively, against D-456 MG. A single dose of 8.1 micromol of intrathecal Spartaject Busulfan resulted in an increase in median survival of 61.7% compared with the saline control. In experiment 2, all busulfan treatments showed increases in median survival of 142.8% (8.1 micromol x 1), 52.3% (2.0 micromol x 4), and 23% (8.1 micromol x 4) (p < 0.001 for all groups) compared with the saline control. These results suggest that a narrow therapeutic dose range for both toxicity and activity has been defined for intrathecal busulfan in the treatment of human neoplastic meningitis in athymic nude rats. Although busulfan has only limited activity against solid tumors, the high doses achievable in the CSF following intrathecal administration coupled with the steep dose-response relationships of alkylating agents, provide rationale for further evaluation of this agent.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Glioblastoma/complications , Glioblastoma/drug therapy , Meningeal Neoplasms/complications , Meningeal Neoplasms/drug therapy , Meningitis/etiology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Busulfan/administration & dosage , Busulfan/adverse effects , Drug Administration Schedule , Female , Injections, Spinal , Rats , Rats, Nude , Subarachnoid Space , Survival Analysis
10.
J Neuroimmunol ; 86(1): 46-52, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9655471

ABSTRACT

We have previously reported that local secretion of either TNF-alpha or TGF beta1 by intracerebral SMA-560 malignant glioma tumor cells can reduce or eliminate tumor growth in mice. However, the use of TNF-alpha, while improving the overall survival of tumor bearing animals, was associated with early toxic deaths due to cerebral edema. In the present study, we demonstrate that TNF-alpha induces apoptosis of the SMA 560 cell line, as does TGF beta1, and that these two cytokines act in an additive fashion to enhance apoptosis and thus, to inhibit SMA 560 cell growth in vitro. Next, we show that the production of TGF beta1 when added to TNF-alpha production by central nervous system tumors in vivo abrogates any early deaths seen due to TNF-alpha toxicity and leads to a larger percentage of animals surviving CNS tumor challenge. Finally, we demonstrate that the production of TGF beta1 by tumor cells is associated with the abolition of tumor-associated cerebral edema in both TNF-alpha and in non-TNF-alpha producing tumors. These results are important for the development of effective and less toxic therapies for brain tumors, as well as for examining the pathogenesis of tumor-related cerebral edema.


Subject(s)
Apoptosis/immunology , Brain Edema/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Transforming Growth Factor beta/biosynthesis , Animals , Apoptosis/drug effects , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Carcinogenicity Tests , Cell Division/drug effects , Disease Models, Animal , Gene Transfer Techniques , Glioma/immunology , Glioma/mortality , Lac Operon , Mice , Mice, Inbred Strains , Retroviridae/genetics , Retroviridae Infections , Survival Analysis , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
J Neurovirol ; 4(2): 148-58, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9584952

ABSTRACT

Any immunotherapeutic approach to cancer cell eradication is based upon the specific recognition of neoplastic cells and the sparing of surrounding normal tissue; perhaps nowhere is this distinction more important than within the central nervous system, due to the diffuse infiltrative nature of primary glial tumor cell growth. Whether ultimate effect moieties are immunoglobulins, fragments and/or their constructs with drugs, toxins, radionuclides, or immune cells, the specificity of effector: cell surface marker is crucial. This review describes the identification, immunologic characterization, and biologic behavior of a transmembrane tumor-specific altered growth factor receptor molecule which may well serve as a mediator of multiple immunotherapeutic approaches: the class III variant of the epidermal growth factor receptor, EGFRvIII.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , ErbB Receptors/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , ErbB Receptors/analysis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioma/chemistry , Glioma/genetics , Glioma/therapy , Humans , Immunohistochemistry , Immunotherapy/methods , Mice , Mice, Nude , Microscopy, Fluorescence , Molecular Sequence Data
12.
Toxicol In Vitro ; 12(4): 483-524, 1998 Aug.
Article in English | MEDLINE | ID: mdl-20654431

ABSTRACT

As a follow-up to a prevalidation study on in vitro tests for replacing the in vivo rabbit test for skin corrosivity, an international validation study was conducted during 1996 and 1997 under the auspices of ECVAM. The main objectives of the study were to: (a) identify tests capable of discriminating corrosives from non-corrosives for selected types of chemicals and/or all chemicals; and (b) determine whether these tests could identify correctly known R35 (UN packing group I) and R34 (UN packing groups II & III) chemicals. The tests evaluated were the rat skin transcutaneous electrical resistance (TER) assay, CORROSITEX(TM), the Skin(2TM) ZK1350 corrosivity test and EPISKIN(TM). Each test was conducted in three independent laboratories. 60 coded chemicals were tested. All of the tests evaluated showed acceptable intralaboratory and interlaboratory reproducibilities, and the TER, Skin(2) and EPISKIN tests proved applicable to testing a diverse group of chemicals of different physical forms, including organic acids, organic bases, neutral organics, inorganic acids, inorganic bases, inorganic salts, electrophiles, phenols and soaps/surfactants. Two of the four tests evaluated, the TER assay and EPISKIN, met the criteria agreed by the Management Team concerning acceptable underprediction and overprediction rates for them to be considered scientifically validated for use as replacements for the animal test for distinguishing between corrosive and non-corrosive chemicals for all of the chemical types studied [objective (a)]. EPISKIN was the only test able to distinguish between known R35 (UN packing group I) and R34 (UN packing groups II & III) chemicals, for all of the chemical types included, on an acceptable number of occasions [objective (b)]. The corrosive potentials of about 40% of the test chemicals could not be assessed with CORROSITEX, and the assay did not meet all of the criteria for it to be considered acceptable as a replacement test. However, CORROSITEX may be valid for testing specific classes of chemicals, such as organic bases and inorganic acids. The Skin(2) assay did not meet the criteria for it to be considered scientifically validated. Thus, the validities of (i) the TER and EPISKIN assays for discriminating corrosives from non-corrosives, and (ii) the EPISKIN assay for identifying correctly known R35/I and R34/II & III chemicals, have been demonstrated in this study. CORROSITEX appears to be valid when used only with certain types of chemicals.

13.
Neurosurgery ; 41(6): 1365-72; discussion 1372-3, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9402588

ABSTRACT

OBJECTIVE: The promise of immunotherapies developed against brain tumors in animal models has not been realized in human clinical trials. This may be because of the routine use of rodent tumors artificially induced by chemicals or viruses that do not accurately portray the intrinsic qualities of spontaneously arising human tumors and that often fail to incorporate the role of immunosuppressants, such as transforming growth factor-beta, that are secreted by human gliomas. From an astrocytoma that arose spontaneously in inbred VM/Dk mice, we have characterized a highly tumorigenic spontaneous murine astrocytoma cell line (SMA-560) that retains features of glial differentiation and naturally produces high levels of biologically active transforming growth factor-beta. We have used this model to determine whether cytokine production by tumor cells will inhibit intracerebral astrocytoma growth. METHODS: Packaging cell lines producing replication-incompetent retroviral vectors were used to transfect the SMA-560 cell line in vitro with the genes encoding the murine cytokines interleukin (IL)-2, IL-3, IL-4, IL-6, tumor necrosis factor-alpha, gamma-interferon, or granulocyte-macrophage colony-stimulating factor or the costimulatory molecule B7.1 (CD80). RESULTS: Mice challenged intracerebrally with 5000 untransfected SMA-560 cells all succumbed to tumor within 30 days, with a median survival of 25 days. In contrast, mice challenged with SMA-560 cells producing IL-2, IL-4, or tumor necrosis factor-alpha each had a more than 400% increase in median survival (P < 0.0001). In these groups, 78.3% (18 of 23 mice), 66.7% (10 of 15 mice), and 60% (6 of 10 mice) of the mice, respectively, remained alive without evidence of tumor for longer than 100 days after the initial tumor challenge. All other cytokines tested and the expression of B7.1 failed to result in an increase in median survival. CONCLUSION: Using a spontaneous astrocytoma model in an inbred mouse strain, we have shown that cytokine production by glial tumors can abrogate their tumorigenicity in vivo despite production of transforming growth factor-beta. These results predict that approaches directed at cytokine production within intracerebral astrocytomas may be efficacious in human trials and that the "immunological privilege" of the brain may not be absolute under such conditions.


Subject(s)
Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cytokines/metabolism , Animals , Carcinogenicity Tests , Female , Genetic Vectors , Histocompatibility Antigens Class I/analysis , Immunohistochemistry , Mice , Mice, Inbred Strains , Retroviridae/genetics , Transfection , Transforming Growth Factor beta/biosynthesis , Tumor Cells, Cultured
14.
J Neuroimmunol ; 78(1-2): 34-46, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9307226

ABSTRACT

An active immunotherapeutic strategy using transfected allogeneic cells for targeting the mutant epidermal growth factor receptor (EGFRvIII) on intracranial tumors was examined. Immunization with allogeneic 300.19/EGFRvIII cells induced CD8+ cytotoxic T-lymphocytes against EGFRvIII bearing syngeneic B16-F10 melanoma or 560 astrocytoma cells (H-2b), but not against allogeneic NR6 cells (H-2q) also bearing EGFRvIII significant NK cell activity was also noted in vitro. Vaccination protected against intracranial challenge with EGFRvIII-positive tumor, with 50% long term survival. In vivo depletions of effector cell subsets demonstrated the requirements for both CD8+ and CD4+ T-cells but not NK cells in producing this protective effect. These data demonstrate the generation of significant, antigen-specific and MHC class I-restricted cytotoxic immune responses which are effective against tumors present in the CNS.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Central Nervous System Neoplasms/prevention & control , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Animals , Antigens, Neoplasm/genetics , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/metabolism , ErbB Receptors/genetics , ErbB Receptors/immunology , Female , Genetic Techniques , Immunization , Mice , Mutation , Neoplasm Transplantation , Survival Analysis , Transfection , Tumor Cells, Cultured
16.
Cancer Res ; 57(8): 1510-5, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9108453

ABSTRACT

Monoclonal antibody (mAb) L8A4, specific for the tumor-associated mutant epidermal growth factor receptor variant III (EGFRvII), is internalized and degraded after cell binding. Four paired-label experiments were performed in athymic mice bearing EGFRvIII-positive xenografts to determine the suitability of N-succinimidyl 3-iodo-5-pyridinecarboxylate (SIPC) for labeling this internalizing mAb. In mice with HC2 20 d2 xenografts, tumor uptake reached a maximum of 32.7 +/- 2.0% injected dose/g when labeled using SIPC, a value significantly higher (P < 0.05, paired t test) than that observed when L8A4 was labeled using lodogen (24.4 +/- 2.2% injected dose/g). The specificity of mAb uptake in HC2 20 d2 and U87MG(delta)EGFR xenografts was measured in separate experiments by coadministration of L8A4 and nonspecific, isotype-matched P3X63Ag8 mAb, both radioiodinated using SIPC. Tumor localization indices were approximately 10 or more by 72 h, a degree of specificity 3-4 times higher than that reported previously when labeling was performed using the tyramine cellobiose (TCB) method. In a final study directly comparing L8A4 labeled using SIPC and TCB, similar tumor levels were obtained (SIPC, 33.7 +/- 6.1% injected dose/g at 24 h; TCB, 37.8 +/- 6.7% injected dose/g at 24 h); however, tumor-to-tissue ratios for the liver, spleen, and kidneys were 3 times higher with SIPC at later time points. These results suggest that SIPC is a promising method for labeling this anti-EGFRvIII mAb and possibly other mAbs that internalize after binding.


Subject(s)
Affinity Labels , Antibodies, Monoclonal/therapeutic use , ErbB Receptors/immunology , Nicotinic Acids/pharmacokinetics , Succinimides/pharmacokinetics , 3T3 Cells/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , ErbB Receptors/metabolism , Mice , Mice, Nude , Tissue Distribution , Transplantation, Heterologous , Tyramine/pharmacokinetics
17.
Cancer Chemother Pharmacol ; 39(4): 307-16, 1997.
Article in English | MEDLINE | ID: mdl-9025771

ABSTRACT

The prognosis for patients with malignant gliomas continues to be dismal. The high degree of resistance of gliomas to nitrosourea-based chemotherapy is one major factor in poor treatment outcome. The identification of O6-alkylguanine-DNA alkyltransferase (AGAT) as a major determinant of nitrosourea resistance has resulted in the development of several agents to inactivate this repair protein and counteract tumor cell resistance. However, a major problem in preclinical trials has been the marked nitrosourea dose limitations imposed by the prior administration of AGAT-depleting agents. We investigated the AGAT depletion and selective enhancement of BCNU activity of intraarterial (i.a.) O6-benzylguanine (O6BG) in the human malignant glioma xenograft D-456 MG growing intracranially (i.e.) in athymic rats. Whereas i.a. O6BG at 2.5 mg/kg produced 100% inhibition of D-456 MG AGAT i.e. activity 8 h after administration, intraperitoneal (i.p.) O6BG at this dose produced only 40% inhibition, requiring dose escalation to 10 mg/kg to produce 100% AGAT depletion. Prior administration of i.p. O6BG (10 mg/kg) and i.a. O6BG (2.5 mg/kg) limited maximum tolerated intravenous (i.v.) BCNU doses (37.5 mg/kg when given alone) to 6.25 and 25 mg/kg, respectively. Higher doses of BCNU alone or in combination with O6BG produced histopathologic evidence of cerebral and hepatic toxicity. Therapy experiments revealed a significantly improved median survival for rats treated with O6BG i.a. (2.5 mg/kg) plus BCNU i.v. (25 mg/kg, days 61 and 59 in duplicate experiments) compared with saline (day 21. P = 0.001). O6BG i.a. or i.p. (days 22 and 23, P = 0.001), BCNU i.v. (37.5 mg/kg, day 29, P = 0.001), and O6BG i.p. (10 mg/kg), plus BCNU i.v. (6.25 mg/kg, day 37, P < 0.001). Therefore, O6BG i.a., by virtue of rapid AGAT depletion and selective uptake into i.c. tumors, offers significant potential for regional chemomodulation of AGAT-mediated nitrosourea resistance in malignant human gliomas with concomitant reduction of systemic toxicity.


Subject(s)
Alkyl and Aryl Transferases , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Carmustine/therapeutic use , DNA Repair , Glioblastoma/drug therapy , Guanine/analogs & derivatives , Transferases/antagonists & inhibitors , Animals , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Guanine/administration & dosage , Guanine/pharmacology , Humans , Injections, Intra-Arterial , Rats , Rats, Nude , Transplantation, Heterologous
18.
Proc Natl Acad Sci U S A ; 93(19): 10399-404, 1996 Sep 17.
Article in English | MEDLINE | ID: mdl-8816812

ABSTRACT

Vaccination with cytokine-producing tumor cells generates potent immune responses against tumors outside the central nervous system (CNS). The CNS, however, is a barrier to allograft and xenograft rejection, and established tumors within the CNS have failed to respond to other forms of systemic immunotherapy. To determine what barriers the "immunologically privileged" CNS would pose to cytokine-assisted tumor vaccines and what cytokines would be most efficacious against tumors within the CNS, we irradiated B16 murine melanoma cells producing murine interleukin 2 (IL-2), IL-3, IL-4, IL-6, gamma-interferon, or granulocyte-macrophage colony stimulating factor (GM-CSF) and used these cells as subcutaneous vaccines against tumors within the brain. Under conditions where untransfected B16 cells had no effect, cells producing IL-3, IL-6, or GM-CSF increased the survival of mice challenged with viable B16 cells in the brain. Vaccination with B16 cells producing IL-4 or gamma-interferon had no effect, and vaccination with B16 cells producing IL-2 decreased survival time. GM-CSF-producing vaccines were also able to increase survival in mice with pre-established tumors. The response elicited by GM-CSF-producing vaccines was found to be specific to tumor type and to be abrogated by depletion of CD8+ cells. Unlike the immunity generated against subcutaneous tumors by GM-CSF, however, the effector responses generated against tumors in the CNS were not dependent on CD4+ cells. These data suggest that cytokine-producing tumor cells are very potent stimulators of immunity against tumors within the CNS, but effector responses in the CNS may be different from those obtained against subcutaneous tumors.


Subject(s)
Brain Neoplasms/immunology , Cancer Vaccines , Cytokines/biosynthesis , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , Animals , Brain Neoplasms/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cell Line , Cytotoxicity, Immunologic , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-3/biosynthesis , Interleukin-4/biosynthesis , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Recombinant Proteins/biosynthesis , Spleen/immunology , Transfection
19.
Nucl Med Biol ; 23(4): 449-58, 1996 May.
Article in English | MEDLINE | ID: mdl-8832699

ABSTRACT

When labeled using the Iodogen method, a chimeric antibody composed of the human IgG2 constant region and the variable regions of murine anti-tenascin 81C6 exhibited superior uptake in human glioma xenografts compared with its murine parent. In the current study, three paired-label experiments were performed in athymic mice with subcutaneous D-54 MG human glioma xenografts to evaluate further the properties of radioiodinated chimeric 81C6. These studies demonstrated that (a) the enhanced tumor uptake of chimeric 81C6 is specific; (b) when labeling was performed using N-succinimidyl 3-iodobenzoate, chimeric 81C6 again showed preferential accumulation in tumor compared with murine 81C6; and (c) the tumor uptake advantage observed previously with murine 81C6 for N-succinimidyl 3-iodobenzoate compared with Iodogen labeling did not occur with chimeric 81C6.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Brain Neoplasms/metabolism , Glioma/metabolism , Immunotoxins/pharmacokinetics , Iodine Radioisotopes , Recombinant Fusion Proteins/pharmacokinetics , Tenascin/immunology , Animals , Humans , Immunoglobulin G/metabolism , Iodine Radioisotopes/pharmacokinetics , Isotope Labeling , Mice , Mice, Nude , Neoplasm Transplantation , Tissue Distribution , Transplantation, Heterologous
20.
Int J Radiat Oncol Biol Phys ; 35(1): 69-80, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8641929

ABSTRACT

PURPOSE: The present study undertook to establish the dose (LD) of systematically administered (via tail vein) sodium [211At]astatide that would kill 10% (LD10) of exposed animals in two mouse models and to evaluate the resulting histologic lesions. METHODS AND MATERIALS: Three dose escalation experiments were carried out using groups of 10 3- to 4-week-old, 20 +/- 2 g B6C3F1 mice, and one dose escalation experiment was carried out with groups of 10 4- to 6-week-old, 22 +/- 2 g BALB/c (nu/nu) mice. All animals were weighed daily and checked twice daily for general health; autopsies were performed within 12 h of death. RESULTS: The LD10 (95% confidence interval) level of free [211At]astatide at 360 days was 15.1 microCi (5.2-19.1 microCi) in B6C3F1 mice and was associated with a 37.8% weight difference from saline controls (p < 0.001). In the BALB/c (nu/nu) mice, the LD10 at 360 days was 7.7 microCi (0-14.2 microCi), while a dose of 10 microCi (0.42 microCi g(-1)) was associated with a 9.44% weight difference vs. saline controls (p < 0.05). Exclusive of the well-known effects on thyroid, [211At]astatide activity levels were associated with severe bone marrow depression, testicular atrophy, focal alopecia, and nuclear atypia of the epidermoid mucosa of the fore-stomach in the B6C3F1 mice; at activity levels approximating LD10 at 360 days, mild changes in the heart, liver, stomach, and spleen were observed. For BALB/c (nu/nu) mice, administration of 10 microCi was associated at autopsy with mild histologic lesions in the heart, stomach, liver, and spleen. CONCLUSIONS: These studies provide a basis for the design of further investigations of [211At]-labeled compounds as therapeutic agents.


Subject(s)
Astatine/toxicity , Animals , Bone Marrow/radiation effects , Heart/radiation effects , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Stomach/pathology , Stomach/radiation effects , Testis/pathology , Testis/radiation effects , Weight Gain/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...