Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Publication year range
1.
Nat Genet ; 2(1): 50-5, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1303250

ABSTRACT

Human mannose binding protein (MBP) is a C-type serum lectin involved in first-line host defense against a variety of bacterial, fungal and viral pathogens. Recently an association was found between low levels of serum MBP and an increased frequency of recurrent infections in infants. A particular genotype, in which glycine is substituted by aspartic acid at codon 54 of MBP in the fifth collagen repeat, shows apparent concordance with the clinical phenotype. We report, however, that this genotype occurs in 5% of the population and encodes a functional protein. Our results indicate that the Gly54Asp allele does not account for a deficiency state, but instead suggest that MBP may have two predominant allelic forms that have overlapping function and differ only in their ability to activate the classical pathway of complement.


Subject(s)
Alleles , Carrier Proteins/genetics , Mannose/metabolism , Base Sequence , Carrier Proteins/blood , Carrier Proteins/immunology , Complement Pathway, Classical , DNA/genetics , Genotype , Humans , Infant , Infections/immunology , Mannose-Binding Lectins , Molecular Sequence Data , Molecular Structure , Opsonin Proteins/genetics , Phenotype , Point Mutation
2.
J Exp Med ; 183(5): 2361-6, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8642346

ABSTRACT

Induction of a T-cell mediated antitumor response is the ultimate goal for tumor immunotherapy. We demonstrate here that antibody-targeted IL2 therapy is effective against established pulmonary and hepatic melanoma metastases in a syngeneic murine tumor model. The effector mechanisms involved in this tumor eradication are not dependent on NK cells, since the therapeutic effect of antibody-IL2 fusion protein was not altered in NK cell-deficient mice. In contrast, T cells are essential for the observed antitumor effect, since therapy with antibody IL2 fusion proteins is unable to induce tumor eradication in T cell-deficient SCID mice. In vivo depletion studies characterized the essential effector cell population further as CD8 + T cells. Such CD8 + T cells, isolated from tumor bearing mice after antibody-directed IL2 therapy, exerted a MHC class I-restricted cytotoxicity against the same tumor in vitro. These data demonstrate the ability of antibody-targeted IL2 delivery to induce a T cell-dependent host immune response that is capable of eradicating established melanoma metastases in clinically relevant organs.


Subject(s)
Interleukin-2/therapeutic use , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphocyte Depletion , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Rats , T-Lymphocyte Subsets/immunology
3.
J Clin Invest ; 98(12): 2801-4, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8981927

ABSTRACT

A major goal of tumor immunotherapy is the induction of tumor-specific T cell responses that are effective in eradicating disseminated tumor, as well as mounting a persistent tumor-protective immunity. We demonstrate here that a genetically engineered fusion protein consisting of human/mouse chimeric anti-ganglioside GD2 antibody and human interleukin-2 is able to induce eradication of established B78-D14 melanoma metastases in immunocompetent syngeneic C57BL/6J mice. This therapeutic effect is mediated by host immune cells, particularly CD8+ T cells and is associated with the induction of a long-lived immunity preventing tumor growth in the majority of animals when challenged up to four months later with B78-D14 cells. This effect was tumor-specific, since no cross-protection against syngeneic, ganglioside GD2+ EL-4 thymoma cells was observed. Furthermore, this tumor-specific protection can be transmitted horizontally to naive, syngeneic SCID mice by passive transfer of CD8+ T lymphocytes derived from immune animals. These results suggest that antibody-targeted delivery of cytokines provides a means to elicit effective immune responses against established tumors in the immunotherapy of neoplastic disease.


Subject(s)
Interleukin-2/therapeutic use , Neoplasms, Experimental/immunology , Recombinant Fusion Proteins/immunology , Animals , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gangliosides/immunology , Immunity , Immunohistochemistry , Immunotherapy , Melanoma/metabolism , Melanoma/therapy , Mice , Mice, SCID , Neoplasm Metastasis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
4.
J Clin Invest ; 105(11): 1623-30, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10841521

ABSTRACT

The induction of tumor-protective immunity against malignancies remains a major challenge in cancer immunotherapy. A novel, humanized anti-ganglioside-GD(2)-IL-2 immunocytokine (hu14.18-IL-2) induced CD8(+) T cells to eradicate established pulmonary metastases of B78-D14 murine melanoma, in a process that required help by CD4(+) T cells and was mediated by the CD40/CD40 ligand (CD40L) interaction. The anti-tumor effect was diminished in mice deficient in CD4(+) T-cells. Three lines of evidence show that CD4(+) T-cell help was mediated by CD40/CD40L interaction but not by endogenous IL-2 production. First, the hu14.18-IL-2-induced anti-tumor response is partially abrogated in C57BL/6J CD40L knockout (KO) mice in contrast to C57BL/6J IL-2 KO animals, in which the immunocytokine was completely effective. Second, partial abrogation of the anti-tumor effect is induced with anti-CD40L antibodies to the same extent as with CD4(+) T-cell depletion. Third, a complete anti-tumor response induced by hu14.18-IL-2 can be reconstituted in C57BL/6J CD40L KO mice by simultaneous stimulation with an anti-CD40 mAb. These results suggest that help provided by CD4(+) T cells via CD40/CD40L interactions in our tumor model is crucial for effective immunotherapy with an IL-2 immunocytokine.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/physiology , Interleukin-2/therapeutic use , Melanoma, Experimental/therapy , Membrane Glycoproteins/physiology , Animals , Antigen-Presenting Cells/physiology , CD40 Antigens/genetics , CD40 Ligand , Female , Lymphocyte Activation , Melanoma, Experimental/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
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
6.
J Natl Cancer Inst ; 93(5): 382-7, 2001 Mar 07.
Article in English | MEDLINE | ID: mdl-11238700

ABSTRACT

BACKGROUND: Angiogenesis is essential for tumor growth and progression. Therefore, inhibition of angiogenesis is being studied as a new anticancer therapy. Because cytotoxic chemotherapy is more effective on rapidly growing tumors than on slowly growing tumors, it has been assumed that antiangiogenic therapy will also be effective only on rapidly growing, highly vascularized tumors. We compared the effects of two angiogenesis inhibitors, TNP-470 and angiostatin, on slowly growing, poorly vascularized and rapidly growing, highly vascularized human tumors in mice. METHODS: Slowly growing (RT-4) and rapidly growing (MGH-U1) human bladder carcinoma cell lines were grown in severe combined immunodeficiency mice. Established tumors were treated with one of the two angiogenesis inhibitors. Tumor volumes, vascularity, and proliferation indices were determined. The in vitro effects of TNP-470 and of angiostatin on the proliferation of RT-4 and MGH-U1 cells were also investigated. All statistical tests were two-sided. RESULTS: RT-4 and MGH-U1 tumor growth was statistically significantly inhibited by both angiogenesis inhibitors (P<.001). Both inhibitors decreased the blood vessel density in both tumor types but did not alter the in vivo proliferation indices of the tumors. TNP-470, but not angiostatin, marginally decreased the in vitro proliferation of MGH-U1 cells. CONCLUSION: Slowly growing, poorly vascularized tumors in animal models respond as well as rapidly growing, highly vascularized tumors to therapy with the angiogenesis inhibitors TNP-470 and angiostatin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Sesquiterpenes/pharmacology , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/drug therapy , Angiostatins , Animals , Carcinoma/blood supply , Carcinoma/drug therapy , Cyclohexanes , Humans , Immunohistochemistry , Mice , Mice, SCID , O-(Chloroacetylcarbamoyl)fumagillol
7.
Cancer Res ; 59(9): 2159-66, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10232603

ABSTRACT

Fusion proteins between whole antibodies (Abs) and cytokines (immunocytokines) such as interleukin 2 have shown efficacy in several mouse tumor models despite a circulating half-life that is significantly shorter than that of the original Ab. We have examined the potential mechanisms responsible for clearance and shown that an important factor is enhanced binding to Fc receptor (FcR). Improvements in the half-lives of two different immunocytokines were made by changing the isotype of the human heavy chain C region from IgG1 or IgG3 to those with reduced binding to FcR, e.g., IgG4. The same effect could also be achieved through site-directed mutagenesis of the FcR binding site in the IgG1 H chain. In vitro studies using mouse J774 FcR-expressing cells showed increased binding of interleukin 2-based immunocytokines, relative to their corresponding Abs, and that this was reversed in those fusion proteins made with IgG4 or mutated IgG1 H chains. All of the fusion proteins showing reduced FcR binding also had reduced Ab-dependent cellular cytotoxicity activity, as measured in 4-h chromium release assays. A complete loss of complement-dependent cytotoxicity activity was seen with an IgG4-based immunocytokine derived from an IgG1 Ab with potent activity. Despite these reduced effector functions, the IgG4-based immunocytokines with extended circulating half-lives showed equivalent (in the case of severe combined immunodeficiency mouse xenograft models) or better (in the case of syngeneic models) efficacy in mouse tumor models than the original IgG1-based molecules. These novel immunocytokines may show improved efficacy in therapeutic situations where T cell- rather than natural killer- or complement-mediated antitumor mechanisms are involved.


Subject(s)
Genes, Immunoglobulin , Immunoconjugates/metabolism , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/metabolism , Interleukin-2/metabolism , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity , Binding Sites , Carcinoma/pathology , Carcinoma/therapy , Complement System Proteins/immunology , Cytotoxicity, Immunologic , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Isotypes/metabolism , Interleukin-2/genetics , Kidney Neoplasms/pathology , Male , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, SCID , Mutagenesis, Site-Directed , Prostatic Neoplasms/pathology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Tissue Distribution , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Cancer Res ; 56(8): 1707-12, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620479

ABSTRACT

Antibody-cytokine fusion proteins can target biologically active cytokines to various tumor sites, achieving local concentrations sufficient to induce host immune responses leading to tumor elimination. Here, we demonstrate the therapeutic efficacy of a tumor-specific antibody-lymphotoxin fusion protein (ch225-LT) on xenografted pulmonary metastases of human melanoma. In vitro studies indicated a direct cytotoxic effect of such construacts on melanoma cells via the induction of apoptosis, as demonstrated by cell cycle analysis and DNA fragmentation. However, ch225-LT lacked any therapeutic effect in immune deficient C.B17 scid/beige and scid/scid mice, indicating the insufficiency of this direct mechanism in vivo. In contrast, in athymic nu/nu mice, ch225-LT completely inhibited outgrowth of the xenografted tumor. This therapeutic effect was accompanied by infiltrations of CD45+, Mac-1+, and asialo-GM1+ cells into the tumor; B220+ cells were present in the surrounding tissue and the periphery of the tumor. The functional role of asialo-GM2+ cells was confirmed by in vivo depletion studies. Our data indicate that an antibody-lymphotoxin fusion protein effectively inhibits the growth of disseminated melanoma metastases by mechanisms that function in the absence of mature T cells, but require B, NK, and other asialo-GM1+ cells.


Subject(s)
B-Lymphocytes/immunology , Immunotoxins/therapeutic use , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphotoxin-alpha/therapeutic use , Melanoma/secondary , Melanoma/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Apoptosis , Cell Cycle , Cell Death , Cell Division/drug effects , DNA Damage , Humans , Immunohistochemistry , Inflammation , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocyte Depletion , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Nude , Mice, SCID , Recombinant Fusion Proteins/therapeutic use , Species Specificity , Transplantation, Heterologous
9.
Cancer Res ; 58(17): 3918-25, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731503

ABSTRACT

The induction of tumor-specific T-cell responses that are effective in eradicating disseminated tumors and in mounting a persistent tumor-protective immunity is one of the major goals of tumor immunotherapy. Here, we demonstrate that we achieved this goal by directing interleukin 2 (IL-2) to the tumor microenvironment of colon carcinoma metastases in syngeneic mice with a recombinant antibody-IL-2 fusion protein (huKS1/4-IL-2). Eradication of established pulmonary metastases is induced by a CD8+ T cell-mediated immune response, which can be transmitted to naive syngeneic severe combined immunodeficient mice by adoptive transfer of CD8+ T cells from immune animals. This immune response was followed by the induction of a long-lived immunity against challenge up to 5 months later with CT26-KSA or wild-type CT26 murine colon carcinoma cells in BALB/c mice. This memory immune response was confirmed by flow cytometric analyses of CD8+ T cells isolated from secondary lymphoid tissue that revealed a phenotypic profile typical of early memory T cells. This long-lived protective tumor immunity was successfully boosted to become optimally effective in all experimental animals by injections of noncurative doses of IL-2 fusion protein 4 days after challenge with tumor cells. Taken together, our results indicate that the huKS1/4-IL-2 fusion protein elicits a long-lived cellular memory immune response that can be amplified by additional applications of IL-2 fusion proteins. This approach could become useful for the treatment of colorectal carcinoma in an adjuvant setting, particularly in patients with minimal residual disease.


Subject(s)
Colonic Neoplasms/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/therapeutic use , Female , Humans , Immunologic Memory , Interleukin-2/therapeutic use , Lung Neoplasms/secondary , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/therapeutic use
10.
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
11.
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
12.
Cancer Res ; 61(4): 1500-7, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245457

ABSTRACT

The huKS1/4-IL2 fusion protein, directed against the human epithelial cell adhesion molecule (huEpCAM) has been shown to induce a strong CD8+ T-cell-dependent, natural killer (NK) cell-independent, antitumor response in mice bearing the huEp-CAM-transfected CT26 colon cancer CT26-EpCAM. Here we investigate the effectiveness of huKS1/4-IL2 against CT26-Ep21.6, a subclone of CT26-EpCAM, expressing low levels of MHC class I. In vitro antibody-dependent cellular cytotoxicity (ADCC) assays in the presence of huKS1/4-IL2 demonstrate that murine NK cells from spleen and blood can kill CT26-Ep21.6 significantly better than they kill CT26-EpCAM. NK-mediated ADCC of CT26-EpCAM can be enhanced by blocking the murine NK cell-inhibitory receptor, Ly-49C. A potent in vivo antitumor effect was observed when BALB/c mice bearing experimental metastases of CT26-Ep21.6 were treated with huKS1/4-IL2. The depletion of NK cells during huKS1/4-IL2 treatment significantly reduced the antitumor effect against CT26-Ep21.6. Together our in vitro and in vivo data in the huEp-CAM-transfected CT26 models indicate that the amount of MHC class I expressed on the tumor target cell plays a critical role in the in vivo antitumor mechanism of huKS1/4-IL2 immunotherapy. A low MHC class I level favors NK cells as effectors, whereas a high level of MHC class I favors T cells as effectors. Given the heterogeneity of MHC class I expression seen in human tumors and the prevailing T-cell suppression in many cancer patients, the observation that huKS1/4-IL2 has the potential to effectively activate an NK cell-based antitumor response may be of potential clinical relevance.


Subject(s)
Adenocarcinoma/immunology , H-2 Antigens/immunology , Immunoconjugates/immunology , Interleukin-2/immunology , Lung Neoplasms/immunology , Adenocarcinoma/secondary , Adenocarcinoma/therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/biosynthesis , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Dose-Response Relationship, Immunologic , Epithelial Cell Adhesion Molecule , Female , H-2 Antigens/biosynthesis , Immunoconjugates/pharmacology , Immunotherapy/methods , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Spleen/cytology , Spleen/immunology , Tumor Cells, Cultured , Up-Regulation
13.
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
14.
Biochim Biophys Acta ; 1088(2): 217-24, 1991 Feb 16.
Article in English | MEDLINE | ID: mdl-1672095

ABSTRACT

We have developed an inducible system for high level expression of heterologous genes in murine hybridoma cells. The rapid induction by methotrexate (MTX) does not involve gene amplification and is controlled at the level of mRNA accumulation. Transfection was achieved by protoplast fusion with an expression vector containing the cDNA of interest and a marker gene encoding dihydrofolate reductase. The initial clones, selected at 100 nM MTX, produced high levels of the protein of interest and contained about 100-400 copies of the integrated plasmid DNA. They could adapt to a 100- to 1000-fold stepwise increase in MTX concentration in a few weeks, during which the expression of the gene of interest but not its copy number, increased several-fold. Furthermore, the induction is freely reversible. If cells were propagated in MTX-free media, the expression level decreased, but the cells could be reinduced to their original high level of expression by adding MTX back to the media. A several-fold increase in the mRNA levels of the dihydrofolate reductase and the gene of interest could be detected after induction for 18 h.


Subject(s)
Methotrexate/pharmacology , Plasminogen Activators/genetics , Poly A/genetics , RNA, Messenger/genetics , Tissue Plasminogen Activator/genetics , Transfection , Urokinase-Type Plasminogen Activator/genetics , Animals , Cell Line , Cloning, Molecular , Gene Expression/drug effects , Genetic Vectors , Humans , Hybridomas , Melanoma , Membrane Fusion , Mice , Plasminogen Activators/metabolism , Restriction Mapping , Tetrahydrofolate Dehydrogenase/genetics , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism
15.
J Clin Oncol ; 16(6): 2169-80, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626218

ABSTRACT

PURPOSE: To evaluate the toxicity, immunogenicity, and pharmacokinetics of a human-mouse chimeric monoclonal antibody (mAb) ch 14.18 directed against disialoganglioside (GD2) and to obtain preliminary information on its clinical efficacy, we conducted a phase I trial in 10 patients with refractory neuroblastoma and one patient with osteosarcoma. PATIENTS AND METHODS: Eleven patients were entered onto this phase I trial. They received 20 courses of mAb ch 14.18 at dose levels of 10, 20, 50, 100, and 200 mg/m2. Dose escalation was performed in cohorts of three patients; intrapatient dose escalation was also permitted. RESULTS: The most prevalent toxicities were pain, tachycardia, hypertension, fever, and urticaria. Most of these toxicities were dose-dependent and rarely noted at dosages of 20 mg/m2 and less. Although the maximum-tolerated dose was not reached in this study, clinical responses were observed. These included one partial (PR) and four mixed responses (MRs) and one stable disease (SD) among 10 assessable patients. Biologic activity of ch 14.18 in vivo was shown by binding of ch 14.18 to tumor cells and complement-dependent cytotoxicity of posttreatment sera against tumor target cells. An anti-ch 14.18 immune response was detectable in seven of 10 patients studied. CONCLUSION: In summary, with the dose schedule used, ch 14.18 appears to be clinically safe and effective, and repeated mAb administration was not associated with increased toxicities. Further clinical trials of mAb ch 14.18 in patients with neuroblastoma are warranted.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Gangliosides/immunology , Neuroblastoma/therapy , Osteosarcoma/therapy , Adult , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Child , Child, Preschool , Complement C3/analysis , Complement C4/analysis , Complement Hemolytic Activity Assay , Female , Fluorescent Antibody Technique, Indirect , Humans , Infusions, Intravenous , Male , Mice , Neuroblastoma/metabolism , Pain/chemically induced , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Treatment Outcome
16.
Pharmacol Ther ; 80(3): 277-92, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9888698

ABSTRACT

Recombinant antibody-cytokine fusion proteins are immunocytokines that achieve high cytokine concentrations in the tumor microenvironment and thereby effectively stimulate cellular immune responses against malignancies. The activation and expansion of immune effector cells, such as CD8+ T lymphocytes, by interleukin-2 immunocytokines resulted in the eradication of established pulmonary and hepatic metastases of murine melanoma and colorectal carcinoma in syngeneic mouse models. These immunocytokines were equally effective in eliminating established bone marrow and liver metastases of murine neuroblastoma by activating natural killer cells. The effective eradication of metastases by immunocytokines resulted in significant prolongation in life span of mice over that of controls receiving equivalent mixtures of antibody and interleukin-2, which failed to reduce the growth of disseminated metastases. Proof of concept was established, indicating that immunocytokine-induced activation and expansion of immune effector cells in the tumor microenvironment can effectively eradicate established tumor metastases. This promising new approach to cancer immunotherapy may lead to clinical applications that improve treatment of cancer patients with minimal residual disease in an adjuvant setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/therapy , Cytokines/therapeutic use , Immunotherapy , Melanoma/therapy , Animals , Colorectal Neoplasms/immunology , Disease Models, Animal , Humans , Melanoma/immunology , Mice
17.
J Mol Biol ; 241(1): 125-7, 1994 Aug 05.
Article in English | MEDLINE | ID: mdl-8051701

ABSTRACT

A trimeric form of the carbohydrate recognition domain of human mannose binding protein has been crystallized in two different forms. The first form crystallizes with symmetry consistent with space group P2(1)2(1)2(1) and a = 61 A; b = 144 A; c = 107 A with presumably two trimers in the asymmetric unit. The second form crystallizes with symmetry consistent with space group P321 and a = b = 77 A; c = 58 A and one monomer per asymmetric unit. The molecular and crystallographic 3-folds must be coincident in this crystal form.


Subject(s)
Carrier Proteins/chemistry , Crystallization , Crystallography, X-Ray , Humans , Mannose-Binding Lectins , Molecular Structure , Molecular Weight , Protein Conformation
18.
Clin Cancer Res ; 7(9): 2862-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11555604

ABSTRACT

PURPOSE: Immune-based therapies, such as the immunocytokine huKS-IL2, exert potent antitumor responses in some animal models by targeting cytokine activity to the tumor microenvironment. We found that certain chemotherapy agents in the appropriate dose and schedule can augment the antitumor activity of huKS-IL2. EXPERIMENTAL DESIGN: Chemotherapy agents were given in a single dose followed 1 day (paclitaxel) or 3 days (cyclophosphamide) later with five daily doses of huKS-IL2 in mice bearing established s.c. tumors, liver metastases, or lung metastases. Tumor models used were CT26/KSA colon, 4T1/KSA mammary, or LLCKSA Lewis lung carcinomas. To measure huKS-IL2 distribution, radiolabeled protein was given to CT26/KSA tumor-bearing mice 1 or 24 h after paclitaxel. huKS-IL2 levels in the tumors were evaluated. RESULTS: Both paclitaxel and cyclophosphamide followed by huKS-IL2 resulted in enhanced antitumor responses compared with either of the treatments alone in the three different tumor models. Results from studies to determine whether the role of the cytotoxic agents in antitumor activity enhancement was related to tumor uptake indicated that a larger fraction of the radiolabeled huKS-IL2 penetrated the tumors when it was administered 24 h after cytotoxic drug "sensitization." CONCLUSION: These data support the idea that prior drug therapy serves to decompress the tumor and lower the diffusion barrier for macromolecules, thus allowing for increased uptake of the huKS-IL2 immunocytokine into the tumor microenvironment. Because the toxicity of the immunocytokine is relatively low at optimal doses, the therapeutic index would likely be greater with the combination treatments.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interleukin-2/immunology , Neoplasms, Experimental/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Cell Survival/drug effects , Cyclophosphamide/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Paclitaxel/administration & dosage , Time Factors , Tumor Cells, Cultured
19.
Clin Cancer Res ; 5(12): 4259-63, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10632368

ABSTRACT

Granulocyte/macrophage-colony stimulating factor (GM-CSF) is very effective at enhancing antibody-dependent cellular cytotoxicity (ADCC) mediated by granulocytes and monocytes. Recently, a fusion protein consisting of GM-CSF and chimeric human/mouse anti-ganglioside G(D2) antibody Ch14.18 (Ch14.18-GM-CSF) has been generated to improve the effectiveness of immunotherapy by directing GM-CSF to the tumor microenvironment and prolonging its relatively short half-life. In this study, we examined the ability of this fusion protein to enhance the in vitro killing of G(D2)-expressing human neuroblastoma cells by granulocytes and mononuclear cells, as well as by complement. The Ch14.18-GM-CSF fusion protein was equally effective as the combination of equivalent amounts of free Ch14.18 and GM-CSF in mediating the killing of NMB7 neuroblastoma cells by granulocytes from seven of eight neuroblastoma patients. The fusion protein was also equally effective as the combination of Ch14.18 and GM-CSF in mediating ADCC by neuroblastoma patients' mononuclear cells. In addition, the fusion protein was as effective as Ch14.18 alone in directing complement-dependent cytotoxicity against NMB7 cells. Our results demonstrate that the biological activities expressed by ADCC and complement-dependent cytotoxicity of both monoclonal antibody Ch14.18 and GM-CSF are retained by the Ch14.18-GM-CSF fusion protein and lend further support for future clinical trials of this fusion protein in patients with neuroblastoma.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Complement System Proteins/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunoconjugates/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cytotoxicity, Immunologic , Gangliosides/immunology , Gangliosides/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocytes/immunology , Humans , Immunoconjugates/immunology , Immunoconjugates/metabolism , Leukocytes, Mononuclear/immunology , Mice , Neuroblastoma/blood , Neuroblastoma/immunology , Neuroblastoma/pathology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
20.
Clin Cancer Res ; 2(12): 1951-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9816154

ABSTRACT

Cytotoxic effector cells interact with target cells through various mechanisms. CTLs use the antigen-specific T cell receptor, whereas Fc receptor-positive natural killer cells use this receptor to interact with antibody-coated target cells. We evaluated the tumor-binding and lymphocyte-activating capability of a recombinant fusion protein consisting of a tumor-selective human/mouse chimeric anti-ganglioside GD2 antibody (ch14.18) and recombinant human interleukin-2 (IL2) (ch14.18-IL2). This fusion protein bound specifically to GD2-positive melanoma and neuroblastoma tumor cell lines, and its IL2 component stimulated in vitro proliferation of an IL2-dependent cell line, as well as peripheral blood mononuclear cells, in healthy control individuals and in cancer patients receiving continuous infusion of IL2. The IL2 presented by the fusion protein, when bound to tumor cells, induced proliferation of IL2-responsive cells as well as a comparable amount of soluble IL2 did. This suggests that localization of IL2 at the site of contact between tumor and effector cells is an effective way of presenting this cytokine to IL2-responsive cells. The ch14.18-IL2 fusion protein also mediated antibody-dependent cellular cytotoxicity with Fc receptor-positive effector cells to an extent similar to ch14.18. These results, together with those of previous studies documenting antitumor efficacy against human tumor xenografts in SCID mice and GD2-positive murine tumors in immunocompetent syngeneic mice, suggest that the ch14.18-IL2 fusion protein should be tested in Phase I and II trials in patients with GD2-positive tumors.


Subject(s)
Gangliosides/immunology , Immunotoxins/therapeutic use , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Animals , Antibodies/genetics , Antibodies/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Division/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunotoxins/genetics , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Mice , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL