Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(23): 8498-503, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731434

ABSTRACT

The induction of a CTL response capable of eradicating disseminated tumor metastases and the establishment of a persistent tumor-protective immunity remain major goals of cancer immunotherapy. Here, we demonstrate for the first time that the combination of interleukin 2 (IL-2) targeted to the tumor microenvironment by a recombinant antibody-IL-2 fusion protein (huKS1/4-IL-2) with gene therapy by the murine chemokine MIG (CXCL9) markedly reduced s.c. tumor burden and decisively suppressed dissemination of experimental lung metastases of CT26-KSA colon carcinoma in syngeneic BALB/c mice. This combined therapy significantly prolonged the life span of these mice 3-4-fold by concurrently delivering MIG and IL-2 to the tumor site and thereby achieving chemoattraction of T cells together with their activation. The antitumor effect obtained was mediated predominantly by MHC class I antigen-restricted CD8(+) T cells with help from MHC class II antigen-restricted CD4(+) T lymphocytes. In addition, the MIG chemokine also induced angiostatic effects in the tumor vasculature. Taken together, this combination of MIG chemokine gene therapy with tumor-targeted cytokine IL-2 provides an approach for the rational design of novel cancer immunotherapy modalities.


Subject(s)
Cell Adhesion Molecules , Chemokines, CXC/genetics , Colonic Neoplasms/therapy , Genetic Therapy/methods , Immunotoxins/pharmacology , Intercellular Signaling Peptides and Proteins , Interleukin-2/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Antigens, Neoplasm/immunology , COS Cells , Cell Division/drug effects , Chemokine CXCL9 , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Combined Modality Therapy , Epithelial Cell Adhesion Molecule , Female , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C
2.
J Immunol ; 167(8): 4560-5, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591784

ABSTRACT

A carcinoembryonic Ag (CEA)-based DNA vaccine encoding both CEA and CD40 ligand trimer achieved effective tumor-protective immunity against murine colon carcinoma in CEA-transgenic mice by activating both naive T cells and dendritic cells. Peripheral T cell tolerance to CEA was broken in a prophylactic model by this novel, dual-function DNA vaccine, whose efficacy was further enhanced by boosts with a recombinant Ab-IL-2 fusion protein (huKS1/4-IL-2). These conclusions are supported by four lines of evidence. First, a lethal challenge of MC38-CEA-KS Ag murine colon carcinoma cells was for the first time completely rejected in 100% of experimental animals treated by oral gavage of this DNA vaccine carried by attenuated Salmonella typhimurium, followed by five boosts with huKS1/4-IL-2. Second, specific activation of dendritic cells was indicated by their marked up-regulation in expression of costimulatory molecules B7.1 (CD80), B7.2 (CD86), and ICAM-1. Third, a decisive increase over control values was observed in both MHC class I Ag-restricted cytotoxicity of CTLs from successfully vaccinated mice and secretion of proinflammatory cytokines IFN-gamma and IL-12. Fourth, activation of CTLs was augmented, as indicated by up-regulation of activity markers LFA-1, CD25, CD28, and CD69. Taken together, these results suggest that a dual-function DNA vaccine encoding CEA and CD40 ligand trimer combined with tumor-targeted IL-2 may be a promising strategy for the rational development of DNA-based cancer vaccines for future clinical applications.


Subject(s)
CD40 Ligand/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/therapeutic use , Carcinoma/prevention & control , Cell Adhesion Molecules , Colonic Neoplasms/prevention & control , Vaccines, DNA/therapeutic use , Animals , Antigens, CD/biosynthesis , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , B7-1 Antigen/biosynthesis , B7-2 Antigen , CD40 Ligand/genetics , CD40 Ligand/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Epithelial Cell Adhesion Molecule , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/therapeutic use , Membrane Glycoproteins/biosynthesis , Mice , Mice, Transgenic , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Vaccination
3.
Vaccine ; 20(3-4): 421-9, 2001 Nov 12.
Article in English | MEDLINE | ID: mdl-11672905

ABSTRACT

A DNA vaccine encoding human carcinoembryonic antigen (CEA) broke peripheral T-cell tolerance toward this tumor self-antigen expressed by Lewis lung carcinoma stably transduced with CEA in C57BL/6J mice transgenic for CEA. This vaccine, delivered by oral gavage with an attenuated strain of Salmonella typhimurium (SL7207), and boosted with an antibody-IL2 fusion protein, induced tumor-protective immunity mediated by MHC class I antigen-restricted CD8(+) T cells, resulting in eradication of subcutaneous tumors in 100% of mice and prevention of experimental pulmonary metastases in 75% of experimental animals. Both CTL and antigen-presenting dendritic cells were activated as indicated by a decisive increase in their respective activation markers CD2, CD25, CD28 as well as CD48 and CD80. The antitumor effects of this CEA-based DNA vaccine obtained in prophylactic settings, suggest that this approach could lead to the rational design of effective treatment modalities for human lung cancer.


Subject(s)
Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Vaccines, DNA/immunology , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Carcinoma, Lewis Lung/prevention & control , Carcinoma, Lewis Lung/secondary , Dendritic Cells/physiology , Histocompatibility Antigens Class I/immunology , Interleukin-12/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vaccination , Vaccines, DNA/administration & dosage
4.
Cancer Res ; 61(16): 6178-84, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507070

ABSTRACT

We demonstrate that a mouse-human chimeric anti-ganglioside GD2-interleukin (IL)-2 fusion protein (ch14.18-IL2) substantially amplifies tumor-protective immunity against murine melanoma induced by an autologous oral DNA vaccine containing the murine ubiquitin gene fused to murine melanoma peptide epitopes gp100(25-35) and TRP-2(181-188). This combination therapy led to the complete rejection of a lethal challenge with B78D14 murine melanoma cells in six of eight mice and a marked suppression of s.c. tumor growth in the two remaining animals. The tumor-protective immunity was mediated by MHC class I antigen- restricted CD8(+) T cells together with CD4(+) T cell help, which was required only for tumor cell killing in the effector phase of the immune response. A single oral vaccination with the DNA vaccine, which was carried by attenuated Salmonella typhimurium, was equally as effective as three such vaccinations applied at 2-week intervals. The immunological mechanisms involved in this antitumor effect were suggested by a decisively increased secretion of tumor necrosis factor alpha TNFTnTNa and IFN-gamma from CD4(+) and CD8(+) T cells and a markedly up-regulated expression on CD8(+) T cells of high-affinity IL-2 receptor alpha chain (CD25), costimulatory molecule CD28, and adhesion molecule lymphocyte function-associated antigen-2 (LFA-2/CD2). Additionally, the combination therapy induced increased expression of costimulatory molecules B7.1 and CD48 on murine antigen-presenting cells. Taken together, our results suggest that IL-2 targeted to the tumor microenvironment by a specific antibody-IL-2 fusion protein is a potent enhancer of tumor-protective immunity induced by an oral DNA vaccine that may ultimately enhance the chances of success in its clinical application.


Subject(s)
Immunotoxins/immunology , Interleukin-2/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Vaccines, DNA/immunology , Administration, Oral , Animals , B7-1 Antigen/biosynthesis , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Drug Synergism , Epitopes/immunology , Female , Gangliosides/immunology , Humans , Immunotoxins/administration & dosage , Interferon-gamma/metabolism , Interleukin-2/administration & dosage , Intramolecular Oxidoreductases/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccines, DNA/administration & dosage , gp100 Melanoma Antigen
5.
Clin Cancer Res ; 7(3 Suppl): 856s-864s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300483

ABSTRACT

Peripheral T-cell tolerance toward human carcinoembryonic self-antigen (CEA) was broken in CEA-transgenic C57BL/6J mice by an oral CEA-based DNA vaccine. This vaccine, delivered by the live, attenuated AroA- strain of Salmonella typhimurium (SL7207), induced tumor-protective immunity mediated by MHC class I-restricted CD8+ T cells. Activation of these T cells was indicated by increased secretion of proinflammatory cytokines IFN-gamma, interleukin (IL)-12 and granulocyte/macrophage-colony stimulating factor, as well as specific tumor rejection and growth suppression in vaccinated CEA-transgenic mice after a lethal challenge with murine MC38 colon carcinoma cells. These tumor cells were double transfected with CEA and the human epithelial cell adhesion molecule (Ep-CAM)/KSA and consequently served as a docking site for a recombinant antibody-IL2 fusion protein (KS1/4-IL2) recognizing KSA. Importantly, the efficacy of the tumor-protective immune response was markedly increased by boosts with this antibody-IL2 fusion protein, resulting in more effective tumor rejection coupled with increased expression of costimulatory molecules B7.2/B7.2 and intercellular adhesion molecule 1 (ICAM-1) on dendritic cells and intensified release of proinflammatory cytokines IFN-gamma, IL-12, and granulocyte/macrophage-colony stimulating factor from T cells of successfully vaccinated CEA-transgenic C57BL/6J mice. Increased T-cell activation mediated by boosts with KS1/4-IL2 fusion protein after tumor cell challenge was further indicated by expanded expression of T-cell activation markers CD25, CD28, CD69, and LFA-1. The application of such CEA-based DNA vaccines and its further improved versions may ultimately prove useful in combination therapies directed against human carcinomas expressing CEA self-antigens.


Subject(s)
Cancer Vaccines , Carcinoembryonic Antigen/metabolism , Vaccines, DNA , Animals , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Colonic Neoplasms/metabolism , Cytokines/metabolism , Epithelial Cell Adhesion Molecule , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunoblotting , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmids/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , Up-Regulation
6.
Protein Expr Purif ; 21(1): 121-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11162396

ABSTRACT

A monoclonal antibody directed against the type 2 adenovirus (Ad2) penton base protein was cloned and expressed in Spodoptera frugiperda (Sf9) cells using a nonlytic vector system. The coding sequences for the immunoglobulin light and heavy chains were placed under the control of the Orgyia pseudotsugata multicapsid nucleopolyhedrosis virus immediate-early 2 (OpIE2) promoter. Transfected Sf9 cells continuously secreted the antibody which retained the ability to recognize both native and recombinant Ad2 penton base proteins. Bifunctional penton base antibodies were also generated by fusing a gene for a growth factor or a cytokine at the 3' end of the Ig constant heavy chain domain. The quantity and activity of recombinant antibodies generated in the nonlytic insect cell system could be determined relatively quickly compared to other expression systems. Moreover, these recombinant proteins were not subjected to proteolytic degradation as frequently occurs during baculovirus-mediated cell lysis and the levels of recombinant antibodies produced in the nonlytic system were comparable to those reported for cytolytic baculovirus vectors.


Subject(s)
Capsid Proteins , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Adenoviridae/immunology , Amino Acid Sequence , Animals , Capsid/immunology , Cell Line , Cloning, Molecular/methods , DNA, Complementary , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Hybridomas , Immunoglobulin G/chemistry , Immunoglobulin G/isolation & purification , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/isolation & purification , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/isolation & purification , Mice , Molecular Sequence Data , Moths , Nucleopolyhedroviruses/genetics , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spodoptera , Transfection/methods
7.
Proc Natl Acad Sci U S A ; 97(10): 5492-7, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10779556

ABSTRACT

We demonstrated that peripheral T cell tolerance toward murine melanoma self-antigens gp100 and TRP-2 can be broken by an autologous oral DNA vaccine containing the murine ubiquitin gene fused to minigenes encoding peptide epitopes gp100(25-33) and TRP-2(181-188). These epitopes contain dominant anchor residues for MHC class I antigen alleles H-2D(b) and H-2K(b), respectively. The DNA vaccine was delivered by oral gavage by using an attenuated strain of Salmonella typhimurium as carrier. Tumor-protective immunity was mediated by MHC class I antigen-restricted CD8(+) T cells that secreted T(H)1 cytokine IFN-gamma and induced tumor rejection and growth suppression after a lethal challenge with B16G3. 26 murine melanoma cells. Importantly, the protective immunity induced by this autologous DNA vaccine against murine melanoma cells was at least equal to that achieved through xenoimmunization with the human gp100(25-33) peptide, which differs in its three NH(2)-terminal amino acid residues from its murine counterpart and was previously reported to be clearly superior to an autologous vaccine in inducing protective immunity. The presence of ubiquitin upstream of the minigene proved to be essential for achieving this tumor-protective immunity, suggesting that effective antigen processing and presentation may make it possible to break peripheral T cell tolerance to a self-antigen. This vaccine design might prove useful for future rational designs of other recombinant DNA vaccines targeting tissue differentiation antigens expressed by tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Melanoma, Experimental/immunology , Vaccines, DNA/therapeutic use , Administration, Oral , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Female , Histocompatibility Antigens Class I/immunology , Humans , Male , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, SCID , Prostatic Neoplasms/immunology , Salmonella typhimurium/immunology , Ubiquitins/genetics , Vaccines, DNA/administration & dosage
8.
Clin Cancer Res ; 4(10): 2551-7, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9796990

ABSTRACT

Immunocytokines are antibody-cytokine fusion proteins that combine the unique targeting ability of antibodies with the multifunctional activities of cytokines to activate effector cells in the tumor microenvironment. Here, we demonstrate the therapeutic efficacy of a tumor-specific immunocytokine, huKS1/4-IL2, which effectively inhibited growth and dissemination of lung and bone marrow metastases of human prostate carcinoma in severe combined immunodeficient mice. This antitumor effect was specific and highly effective, irrespective of reconstitution of these mice with human lymphokine-activated killer cells. Survival times of mice treated with huKS1/4-IL2 were increased 4-fold as compared with animals treated with a mixture of the corresponding antibody and recombinant human interleukin-2 (rhIL2). A persistent antitumor response after treatment with the huKS1/4-IL2 immunocytokine in B, T, and natural killer cell-deficient severe combined immuodeficient-BEIGE mice, depleted of granulocytes, implies a major role for macrophages in this treatment effect. Our data demonstrate that immunocytokine-directed interleukin-2 therapy to tumor sites is an immunotherapeutic approach with potent effects against disseminated metastases of human prostate carcinoma and suggest that this treatment could be effective in an adjuvant setting for patients with minimal residual disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-2/therapeutic use , Prostatic Neoplasms/therapy , Animals , Cell Adhesion Molecules/immunology , Humans , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Male , Mice , Mice, SCID
9.
Bioconjug Chem ; 9(4): 482-9, 1998.
Article in English | MEDLINE | ID: mdl-9667950

ABSTRACT

Localized cytokine therapies with recombinant monoclonal antibody-cytokine fusion proteins, designated immunocytokines, have become of increasing interest for tumor immunotherapy, since they direct immunomodulatory cytokines into the tumor microenvironment. To investigate their mechanisms of action in a variety of syngeneic tumor models, recombinant mouse cytokines IL2 and GM-CSF were engineered as fusion proteins to the carboxyl terminus of a chimeric rat/mouse antitransferrin receptor antibody, ch17217 and expressed in stable-transfected Chinese hamster ovary cells. The recombinant immunocytokines were readily purified by affinity chromatography and their binding characteristics were identical to those shown for the ch17217 antibody. The IL2 immunocytokine had an activity similar to recombinant mouse IL2, whereas the GM-CSF immunocytokine had enhanced cytokine activity relative to recombinant mouse GM-CSF. The clearance rates of ch17217 and the GM-CSF and IL2 immunocytokines were relatively similar with elimination phases (t1/2alpha) of 1.8 h and distribution phases (t1/2beta) of 83, 88, and 91 h, respectively. Both immunocytokines demonstrated effective antitumor activity by suppressing the growth of hepatic metastases of mouse neuroblastoma and pulmonary metastases of mouse colon carcinoma in syngeneic A/J and BALB/c mice, respectively. These results indicate that biologically effective IL2 and GM-CSF immunocytokines combine the targeting ability of an antitransferrin receptor monoclonal antibody with the immunomodulatory functions of each cytokine. Because of the universal expression of the transferrin receptor on mouse tumor cell lines, these constructs should prove useful to determine their efficacy in a wide variety of syngeneic mouse tumor models and to perform detailed studies of their modes of action.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunotoxins/chemistry , Immunotoxins/pharmacology , Interleukin-2/chemistry , Interleukin-2/pharmacology , Receptors, Transferrin/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , CHO Cells/metabolism , Colonic Neoplasms/therapy , Colonic Neoplasms/ultrastructure , Cricetinae , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Immunotoxins/pharmacokinetics , Interleukin-2/pharmacokinetics , Mice , Mice, Inbred A , Mice, Inbred BALB C , Neuroblastoma/therapy , Neuroblastoma/ultrastructure , Rats , Receptors, Transferrin/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
10.
Cancer Res ; 57(21): 4948-55, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9354462

ABSTRACT

A recombinant humanized antibody-interleukin 2 fusion protein (huKS1/4-IL-2) was used to direct IL-2 to the tumor microenvironment and elicit a T cell-mediated eradication of established pulmonary and hepatic CT26-KSA colon carcinoma metastases in syngeneic BALB/c mice. This antitumor effect was specific because a fusion protein, which was nonreactive with these tumor cells, failed to exert any such effect. The efficacy of the huKS1/4-IL-2 fusion protein in eliminating metastases was documented because mixtures of monoclonal antibody huKS1/4 with recombinant human IL-2 were ineffective and, at best, only partially reduced tumor load. Two lines of evidence indicated the eradication of metastases and the absence of minimal residual disease in animals treated with the fusion protein: first, the lack of detection of CT26-KSA cells by reverse transcription-PCR, which can detect one tumor cell in 10(6) liver cells; and second, the tripling of life span. The effector mechanism involved in this tumor eradication is dependent on T cells because the IL-2-directed therapy is ineffective in T cell-deficient SCID mice. The essential effector cells were further characterized as CD8+ T cells by in vivo depletion studies. Such T cells, isolated from tumor-bearing mice after fusion protein therapy, elicited MHC class I-restricted cytotoxicity in vitro against colon carcinoma target cells. Taken together, these data indicate that fusion protein-directed IL-2 therapy induces a T cell-dependent host immune response capable of eradicating established colon cancer metastases in an animal tumor model.


Subject(s)
Cell Adhesion Molecules , Colonic Neoplasms/pathology , Immunotoxins/therapeutic use , Interleukin-2/therapeutic use , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Antibodies/therapeutic use , Antigens, Neoplasm/analysis , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Epithelial Cell Adhesion Molecule , Female , Humans , Immunity, Cellular , Immunotoxins/pharmacokinetics , Interleukin-2/pharmacokinetics , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Recombinant Fusion Proteins/pharmacokinetics , T-Lymphocyte Subsets/immunology , Tumor Cells, Cultured
11.
J Natl Cancer Inst ; 89(21): 1586-94, 1997 Nov 05.
Article in English | MEDLINE | ID: mdl-9362156

ABSTRACT

BACKGROUND: Advanced (stage 4) cases of neuroblastoma, a childhood cancer of the nervous system, are associated with high relapse rates, even after intensive chemotherapy, radiotherapy, and autologous bone marrow transplantation. Therefore, the use of monoclonal antibodies directed against the neuroblastoma tumor marker disialoganglioside GD2 (GD2), in combination with recombinant human interleukin 2 (rhIL-2), is under clinical investigation. We hypothesize that targeted cytokine immunotherapy with a recombinant anti-GD2 antibody-interleukin 2 fusion protein (ch14.18-IL-2) is superior to a combination of ch14.18 and rhIL-2. Our purpose was as follows: 1) to develop a syngeneic model for murine neuroblastoma that expresses GD2 and features both experimental and spontaneous metastases to bone marrow and liver, and 2) to assess anti-GD2-targeted IL-2 therapy in this mode. METHODS: A hybrid neuroblastoma cell line was used to generate the GD2-positive NXS2 cell line. Bone marrow and liver metastases were quantified by reverse transcription-polymerase chain reaction for tyrosine hydroxylase and by organ weight or counts of macroscopic tumor foci, respectively. All P values reported are two-sided. RESULTS: Injection of NXS2 cells resulted in disseminated bone marrow and liver metastases exhibiting stable, but heterogeneous expression of GD2. Treatment with fusion protein (10 microg/day for 6 days) effectively suppressed growth of both experimental and spontaneous metastases to bone marrow and liver (P<.001). In contrast, a mixture of rhIL-2 and ch14.18 at equivalent dose levels was inefficient. Only mice treated with ch14.18-IL-2 showed a twofold prolongation in life span (P<.001). CONCLUSION: Targeted IL-2 therapy with a ch14.18-IL-2 fusion protein elicits an effective antitumor response. Our data suggest that a study of ch14.18-IL-2 as an adjuvant treatment in patients with minimal residual disease may be of value.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Marrow Neoplasms/therapy , Interleukin-2/therapeutic use , Neuroblastoma/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor/immunology , Bone Marrow Neoplasms/immunology , Bone Marrow Neoplasms/secondary , Female , Gangliosides/immunology , Incidence , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Mice , Neoplasm Metastasis , Neuroblastoma/immunology , Neuroblastoma/secondary , Polymerase Chain Reaction/methods , Transcription, Genetic , Tumor Cells, Cultured
12.
J Appl Physiol (1985) ; 82(5): 1445-52, 1997 May.
Article in English | MEDLINE | ID: mdl-9134891

ABSTRACT

We investigated methacholine (MCh)-induced bronchoconstriction in explanted airways from Fischer and Lewis rats. Lung explants, 0.5- to 1.0-mm thick, were prepared from agarose-inflated lungs of anesthetized 8- to 12-wk-old male rats. After overnight culture, videomicroscopy was used to record baseline images of the individual airways. Dose-response curves to MCh were then constructed by repeated administration of MCh; airways were reimaged 10 min after each MCh administration. Airway internal luminal area (Ai) was measured at successive MCh concentrations from 10(-9) to 10(-1) M. In addition to the effective concentration leading to 50% of the achieved maximal response, we also determined the effective concentration leading to a 40% reduction in Ai. Both the effective concentration leading to 50% of the achieved maximal response and the concentration leading to a 40% reduction in Ai were significantly lower among Fischer rat airways (P < 0.05). Airway closure was more common among Fischer rat airways (17%) than among those of Lewis rats (7.5%). Responsiveness of Fischer rat airways was more heterogeneous than among Lewis airways; a larger number of Fischer rat airways exhibited high sensitivity to MCh. There was no relationship between responsiveness and baseline Ai in either strain. In a second experiment, we measured the rate of contraction of explanted airways from lungs inflated to 50, 75, and 100% of total lung capacity. The average rate of contraction in the first 15 s was higher in Fischer rat airways at each inflation volume. These data indicate that the hyperresponsiveness of the Fischer rat reflects the responsiveness of individual airways throughout the airway tree and are consistent with the notion that in this model hyperresponsiveness is an intrinsic property of airway smooth muscle.


Subject(s)
Bronchoconstriction/physiology , Animals , Bronchi/drug effects , Bronchi/physiology , Bronchoconstriction/drug effects , Bronchoconstrictor Agents/pharmacology , Dose-Response Relationship, Drug , Male , Methacholine Chloride/pharmacology , Muscle, Smooth/physiology , Organ Culture Techniques , Population , Rats , Rats, Inbred F344 , Rats, Inbred Lew
13.
Lung ; 175(3): 155-63, 1997.
Article in English | MEDLINE | ID: mdl-9087943

ABSTRACT

Pretreatment with inhaled fuorsemide has been shown to protect against bronchoconstrictive stimuli that indirectly activate airway smooth muscle. However, it is controversial as to whether furosemide acts directly on airway smooth muscle. To investigate this we studied the effect of furosemide on both methacholine (MCh)- and serotonin (5-HT)-induced bronchoconstriction in explanted rat airways. Lungs from 21 Sprague-Dawley rats (269 +/- 15 g) were excised, inflated with agarose solution at 37 degrees C (1% w/v, 48 ml/kg), embedded in 4% agarose, and refrigerated to gel the agarose. Lung slices (0.5-1.0 mm thick) were cultured overnight at 37 degrees C. Explants were placed on a dissecting video microscope, and airway area was measured with an image analysis system. MCh or 5-HT was administered directly to explanted airways (final concentrations 3.8 x 10(-6) M and 3.8 x 10(-5) M, respectively). Five min later furosemide (3.7 x 10(-5) M or 3.7 x 10(-4) M) was added and airway area monitored 5, 10, 15, 30, and 60 min later. Results were expressed as a percentage of the maximal response. Significant bronchodilation was seen after 30 min in airways preconstricted with MCh and after 15 min in those preconstricted with 5-HT following 3.7 x 10(-4) M furosemide (p < 0.05). 3.7 x 10(-5) M furosemide caused bronchodilation only at 60 min in airways constricted with 5-HT. The effect was blocked by a 30-min incubation of explants with 10(-6) M indomethacin. The furosemide-induced bronchodilation effect was not observed in airways strongly constricted with 3.8 x 10(-5) M MCh. These findings indicate that in the rat at least, furosemide induces a weak bronchodilator effect present only at high doses, which seems to be dependent on the production of prostaglandins. This effect may be relevant to the observed therapeutic action of furosemide in asthmatics.


Subject(s)
Bronchi/drug effects , Bronchoconstriction/drug effects , Bronchodilator Agents/pharmacology , Diuretics/pharmacology , Furosemide/pharmacology , Prostaglandins/biosynthesis , Animals , Bronchi/metabolism , Bronchi/physiology , Bronchoconstriction/physiology , Bronchoconstrictor Agents/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Indomethacin/pharmacology , Male , Methacholine Chloride/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Time Factors
14.
J Comp Psychol ; 110(1): 45-54, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8851552

ABSTRACT

This study compared the direction of social learning in 2 populations of Barbados Zenaida doves (Zenaida aurita). One population (St. James) is territorial; it competes aggressively with conspecifics but scramble competes with heterospecifics. The other population (Deep Water Harbour) forages in large homospecific flocks. Field observations were conducted to quantify intraspecific and interspecific patterns of foraging association and aggression. Wild-caught doves from both areas were then tested on novel foraging tasks demonstrated by either a conspecific or a heterospecific tutor. In all experiments, St. James doves learned more readily from the heterospecific tutor (Carib grackle -Quiscalus lugubris-), whereas Deep Water Harbour doves learned more readily from the conspecific tutor. The type of competitive feeding interaction in the field (i.e., scramble vs. interference) appears to better predict the pattern of social learning in an experiment than does species identity.


Subject(s)
Appetitive Behavior , Columbidae , Competitive Behavior , Imitative Behavior , Social Environment , Aggression/psychology , Animals , Barbados , Female , Male , Species Specificity , Territoriality
SELECTION OF CITATIONS
SEARCH DETAIL
...