Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 93(4): 556-65, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11477560

ABSTRACT

1D10 is a previously described antibody that binds to cells from a majority of B-cell malignancies. The current studies were designed to further evaluate the antigen specificity of 1D10 and its potential as an immunotherapeutic agent. Studies with transfectants and immunoprecipitation demonstrated that 1D10 recognizes some, but not all, of the human HLA-DR beta chains. Both normal and malignant B cells can express the 1D10 antigen. A humanized version of 1D10 was produced using CDR grafting. The resulting antibody has an affinity that is similar to that of the parental murine antibody. In addition, the humanized antibody is capable of inducing complement-mediated cytotoxicity, antibody-dependent cell cytotoxicity, and direct apoptosis of 1D10-expressing B cells. Based on these in vitro anti-tumor activities, we conclude humanized 1D10 deserves further evaluation as an immunotherapeutic agent.


Subject(s)
Antibodies, Monoclonal/immunology , HLA-DR Antigens/immunology , Immunoglobulin G/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibody Affinity , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity , Apoptosis/immunology , Cloning, Molecular , Epitopes/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Tumor Cells, Cultured
2.
Transplantation ; 71(7): 941-50, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11349730

ABSTRACT

BACKGROUND: Humanized Fc receptor (FcR)-nonbinding antibodies against CD3 are promising immunosuppressive agents that may overcome both the neutralizing response to and the cytokine release syndrome seen with conventional monoclonal antibodies against CD3. In addition, evidence from several murine models suggests that these recombinant antibodies may actively induce T cell unresponsiveness by a mechanism other than modulation of the T cell receptor (TCR) or T cell depletion. We hypothesized that FcR-nonbinding antibodies against CD3 could induce T cell unresponsiveness by acting as partial agonist ligands of the TCR and thus, inducing T cell anergy. METHODS: To test this hypothesis, we examined the signaling and functional effects of HuM291 (Nuvion), a FcR-nonbinding humanized antibody against CD3, on primary human T cells. RESULTS: Short exposure of human peripheral blood T lymphocytes to HuM291 caused a partial agonist type of signaling through the TCR characterized by incomplete phosphorylation of TCR zeta, failure to activate ZAP-70 and to phosphorylate LAT but activation of ERK-1/-2 and subsequent up-regulation of CD69 expression. These changes correlated with a dose-dependent induction of anergy in human, primary resting T cells, which was reversed by exogenous interleukin-2. CONCLUSIONS: The tolerogenic properties of FcR-nonbinding monoclonal antibodies against CD3 correlate with its ability to reproduce the biochemical and functional effects of TCR partial agonist ligands. Thus, generation of engineered antibodies against CD3 with low TCR oligomerization potential may provide a clinically applicable partial agonist-based strategy for the prevention of polyclonal T cell responses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , Clonal Anergy , Receptors, Antigen, T-Cell/agonists , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Blood Cells/drug effects , Blood Cells/immunology , Humans , Receptors, Fc/immunology
5.
J Immunol ; 165(11): 6205-13, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11086054

ABSTRACT

Human trials in organ allografts have demonstrated that murine anti-CD3 mAbs are immunosuppressive. By mimicking Ag, anti-CD3 can produce T cell activation, anergy, or death. Activation of resting T cells in vivo results in dose-limiting cytokine release and is caused by Ab-mediated cross-linking of T cells and Fcgamma receptor (FcR)-bearing cells. With the goal of minimizing cytokine-induced toxicity, anti-CD3 have been engineered to lower Fc binding avidity. Preclinical murine studies have indicated that non-FcR-binding anti-CD3 can induce apoptosis of Ag-activated T cells. Since induction of T cell apoptosis may be an important mechanism of immunosuppression by anti-CD3, we tested whether Fc mutations affect the ability of anti-human CD3 to induce apoptosis of activated T cells. We compared wild-type murine anti-CD3, M291, and OKT3 and their humanized, FcR- and non-FcR-binding structural variants in quantitative assays of T cell apoptosis. Non-FcR-binding variants produced more sustainable phosphorylation of extracellular signal-regulated kinase-2, greater release of IFN-gamma, and more effectively caused activation-dependent T cell apoptosis. Non-FcR-binding variants dissociated more quickly from the T cell surface and caused less internalization of the TCR, which then remained available in greater abundance on the cell surface for signaling. Cross-linking of non-FcR-binding variants by antiglobulin enhanced TCR internalization and minimized induction of T cell apoptosis. We conclude that non-FcR-binding, humanized anti-CD3 have improved ability to induce apoptosis of activated T cells, presumably by allowing durable expression of the TCR and sustained signaling.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/immunology , Binding Sites, Antibody , CD3 Complex/immunology , Lymphocyte Activation/immunology , Receptors, Fc/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Adult , Animals , Antibodies, Monoclonal/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle/immunology , Cells, Cultured , Down-Regulation/immunology , Humans , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Muromonab-CD3/metabolism , Muromonab-CD3/pharmacology , Phosphorylation , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell/metabolism , Solubility , T-Lymphocyte Subsets/metabolism
6.
Cancer Res ; 60(24): 6977-84, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11156399

ABSTRACT

Adult T-cell leukemia (ATL) develops in a small proportion of human T-cell lymphotrophic virus-I infected individuals. The leukemia consists of an overabundance of activated T cells, which are characterized by the expression of CD25, or IL-2Ralpha, on their cell surface. Presently, there is not an accepted curative therapy for ATL. We developed an in vivo model of ATL in non-obese diabetic/severe combined immunodeficient (NOD/ SCID) mice by introducing cells from an ATL patient (MET-1) into the mice. The leukemic cells proliferated in these mice that lack functional T, B, and natural killer (NK) cells. The MET-1 leukemic cells could be monitored by measurements of both serum soluble Tac (IL-2Ralpha) and soluble human beta2-microglobulin (beta2mu) by ELISA. The disease progressed to death in the mice after approximately 4-6 weeks. The mice developed grossly enlarged spleens and a leukemia involving ATL cells that retained the phenotype and the T-cell receptor rearrangement and human T-cell lymphotrophic virus-I integration pattern of the patient's ATL leukemia cells. This model is of value for testing the efficacy of novel therapeutic agents for ATL. The administration of humanized anti-Tac (HAT), murine anti-Tac (MAT), and 7G7/B6, all of which target IL-2Ralpha, significantly delayed the progression of the leukemia and prolonged the survival of the tumor-bearing mice. In particular, HAT induced complete remissions in 4 of 19 mice and partial remissions in the remainder. It appears that the antibodies act by a mechanism that had not been anticipated. The prevailing view is that antibodies to the IL-2Ralpha receptor have their effective action by blocking the interaction of IL-2 with its growth factor receptor, thereby inducing cytokine deprivation apoptosis. However, although both HAT and MAT block the binding of IL-2 to IL-2Ralpha of the high affinity receptor, the 7G7/B6 monoclonal antibody binds to a different epitope on the IL-2Ralpha receptor, one that is not involved in IL-2 binding. This suggested that the antibodies provide an effective therapy by a mechanism other than induction of cytokine deprivation. In accord with this view, the MET-1 cells obtained from the spleens of leukemic mice did not produce IL-2, nor did they express IL-2 mRNA as assessed by reverse transcription-PCR. Another possible conventional mechanism of action involves complement-mediated killing. However, although MAT and 7G7/B6 fix rabbit complement, HAT does not do so. Furthermore, in the presence of NOD/SCID mouse serum, there was no complement-mediated lysis of MET-1 cells. In addition, the antibodies did not manifest antibody-dependent cellular cytotoxicity with NOD/SCID splenocytes that virtually lack NK cells as the effector cells as assessed in an in vitro chromium-release assay. However, in contrast to the efficacy of intact HAT, the F(ab')2 version of this antibody was not effective in prolonging the survival of mice injected with MET-1 ATL cells. In conclusion, in our murine model of ATL, monoclonal antibodies, HAT, MAT, and 7G7/B6, appear to delay progression of the leukemia by a mechanism of action that is different from the accepted mechanism of IL-2 deprivation leading to cell death. We consider two alternatives: the first, antibody-dependent cellular cytotoxicity mediated by FcRI- or FcRIII-expressing cells other than NK cells, such as monocytes or polymorphonuclear leukocytes. The second alternative we consider involves direct induction of apoptosis by the anti-IL-2R antibodies in vivo. It has been shown that the IL-2R is a critical element in the peripheral self-tolerance T-cell suicide mechanism involved in the phenomenon of activation-induced cell death.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Leukemia, T-Cell/therapy , Receptors, Interleukin-2/immunology , Animals , B-Lymphocytes/immunology , Complement System Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Humans , Killer Cells, Natural/immunology , Leukemia/immunology , Leukemia/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Receptors, Interleukin-2/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Time Factors , beta 2-Microglobulin/blood
7.
Transplantation ; 68(4): 563-71, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10480417

ABSTRACT

BACKGROUND: OKT3, a mouse monoclonal antibody (Ab) specific for the human CD3 complex on T cells, is a potent immunosuppressive agent used for the treatment of acute allograft rejection. The utility of the drug has been limited by a neutralizing anti-mouse Ab response and adverse side effects resulting from T cell activation and systemic cytokine release. T cell activation is caused by OKT3-mediated cross-linking of T cells and Fc receptor-bearing cells. Studies in the mouse model have shown that global T cell activation is not necessary for immunosuppression, as Fc receptor-nonbinding anti-CD3 Abs can suppress graft rejection in the absence of the activation effects seen with Fc receptor-binding Abs. Thus, a humanized anti-CD3 antibody with a low affinity for Fc receptors might improve immunosuppressive therapy by reducing the side effects associated with OKT3. METHODS: We developed a mouse monoclonal Ab, M291, which competes with OKT3 for binding to T cells. Humanized, complementary-determining region-grafted versions of M291 featuring various Fc were engineered, including a previously described IgG2 mutant deficient in Fc receptor binding (HuM291). RESULTS: Compared with OKT3 and HuM291-IgG1, HuM291 was significantly less mitogenic to T cells in vitro and induced the release of much lower levels of the cytokines tumor necrosis factor-alpha, interferon-gamma, and interleukin-10. Despite this reduction in T cell activation, HuM291 retained the ability to modulate the CD3 complex and inhibit the mixed lymphocyte reaction. CONCLUSIONS: When evaluated in vivo, HuM291 may be an immunosuppressive agent associated with less of the acute toxicity and immunogenicity seen with OKT3 therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antilymphocyte Serum/pharmacology , CD3 Complex , Immunosuppressive Agents/pharmacology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/genetics , Antibody Affinity , Antibody Specificity , Antilymphocyte Serum/adverse effects , Antilymphocyte Serum/genetics , Cytokines/biosynthesis , DNA, Complementary/genetics , Drug Design , Humans , Immunoglobulin Variable Region/genetics , In Vitro Techniques , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mitogens/pharmacology , Molecular Sequence Data , Muromonab-CD3/adverse effects , Muromonab-CD3/pharmacology , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/cytology
8.
Int J Cancer ; 77(2): 251-6, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9650561

ABSTRACT

Anti-CD3 x anti-B-cell antigen bispecific monoclonal antibodies (bsAbs) can redirect T-cell-mediated lysis toward malignant B cells. Clinical trials with CD3-based bsAbs have shown toxicity in patients which is likely related to nonspecific T-cell activation and targeting. Our current studies were designed to explore the mechanisms responsible for the observed in vivo toxicity by evaluating the immunologic effects of 2 different bsAb preparations in vitro. 1D10 was used as the tumor specific arm of the bsAbs. This antibody reacts with a variant of HLA-DR found on a majority of pre-B- and B-cell malignancies, and normal B cells in some individuals. Anti-CD3 served as the T-cell specific arm. A 1D10 x anti-CD3 bispecific IgG (bsIgG) produced using the hybrid-hybridoma method was compared to a 1D10 x anti-CD3 bispecific F(ab')2 [bsF(ab')2] produced using the leucine zipper technique. In cytotoxicity assays, both bsIgG and bsF(ab')2 induced lysis by pre-activated T cells of 1D10 (+) malignant B cells. bsIgG at high concentrations also induced lysis of 1D10 (-) tumor cells, while bsF(ab')2 did not. Proliferation of T cells induced by bsIgG and bsF(ab')2 was also evaluated. Both forms of bsAbs induced T-cell proliferation in the presence of antigen (+) Raji cells, while only bsIgG did so in the presence of antigen (-) malignant B cells. bsF(ab')2 induced T-cell activation in the absence of any tumor cells when testing was performed on samples where the 1D10 target antigen was present on normal peripheral blood B cells. We conclude that non-specific T-cell activation from bsAbs can occur in an antigen-independent manner due to the Fc/Fc receptor (FcR) interaction, or in an antigen-dependent manner when antigen is expressed on normal or tumor cells. Both mechanisms may have been responsible for the toxicity observed in prior clinical studies.


Subject(s)
Antibodies, Bispecific/immunology , B-Lymphocytes/immunology , CD3 Complex/immunology , Immunoglobulin Fab Fragments/immunology , Receptors, Fc/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , Cytotoxicity, Immunologic , Humans , Immunoglobulin G/immunology , Jurkat Cells , Lymphocyte Activation , Mice , Tumor Cells, Cultured
9.
J Exp Med ; 187(8): 1249-60, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9547336

ABSTRACT

The signals that direct differentiation of T cells to the CD4 or CD8 lineages in the thymus remain poorly understood. Although it has been relatively easy to direct differentiation of CD4 single positive (CD4+) cells using combinations of antibodies and pharmacological agents that mimic receptor engagements, equivalent stimuli do not induce efficient maturation of CD8+ cells. Here we report that, irrespective of the MHC-restriction specificity of the TCR, differentiation of mature CD8+ thymocytes can be induced by ligation of CD3 polypeptides on immature thymocytes with a F(ab')2 reagent (CD3fos-F(ab')2). The tyrosine phosphorylation patterns stimulated by CD3fos-F(ab')2 have been shown to resemble those delivered to mature T cells by antagonist peptides, which are known to direct positive selection of CD8+ cells, and we can show that this reagent exhibits potent antagonistic-like activity for primary T cell responses. Our results suggest a distinction in the signals that specify lineage commitment in the thymus. We present a model of thymocyte differentiation that proposes that the relative balance of signals delivered by TCR engagement and by p56lck activation is responsible for directing commitment to the CD8 or CD4 lineages.


Subject(s)
CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cells/immunology , Receptors, Antigen, T-Cell/immunology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Lineage , Cytotoxicity Tests, Immunologic , Histocompatibility Antigens , Immunoglobulin Fragments/immunology , Immunologic Capping , Ligands , Lymphocyte Activation , Mice , Mice, Mutant Strains , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Thymus Gland/cytology , Tyrosine/metabolism , ZAP-70 Protein-Tyrosine Kinase
10.
J Immunol ; 159(7): 3613-21, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9317161

ABSTRACT

The mouse anti-human CD3 mAb OKT3 is a potent immunosuppressive agent used for the treatment of acute transplant rejection. OKT3 therapy is associated with acute toxicity resulting from in vivo T cell activation and systemic cytokine release, and a human anti-mouse Ab response. T cell activation is thought to be triggered by CD3 cross-linking mediated by the Abs bridging T cells and Fc receptor-bearing cells. Recent studies in a mouse model indicate that anti-mouse CD3 Abs with low affinity for Fc receptors can achieve immunosuppression without T cell activation, toxicity, or an anti-Ab response. To obtain an analogous Ab to improve the current anti-human CD3 therapy, a humanized Ab with low affinity for Fc receptors is needed. In this study, we introduced mutations into the upper CH2 region of IgG2 and expressed the altered Fc as chimeric OKT3 Abs. Compared with chimeric OKT3 IgG1, IgG2, IgG3, and IgG4, the IgG2 mutants were less mitogenic to T cells, and they did not induce the release of TNF-alpha, IFN-gamma, or IL-2. In parallel, we observed no functional interaction of the IgG2 mutant Abs with K562 cells, which express the IgG2-binding Fc receptor on their surface. Despite no measurable T cell activation, the mutant Abs could still modulate the CD3 complex. When coupled to a humanized anti-CD3, the IgG2 variant may provide a drug with less acute toxicity and immunogenicity, but may still retain potent immunosuppressive properties.


Subject(s)
Immunoglobulin G/genetics , Lymphocyte Activation , Mitogens/pharmacology , Muromonab-CD3/pharmacology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibody Affinity , CD3 Complex/immunology , Cytokines/pharmacokinetics , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/physiology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Isotypes/genetics , Lymphocyte Activation/drug effects , Mice , Mitogens/immunology , Muromonab-CD3/genetics , Muromonab-CD3/metabolism , Mutagenesis, Site-Directed/immunology , Receptors, IgG/physiology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/drug effects
11.
J Immunol ; 159(3): 1543-56, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9233654

ABSTRACT

Humanized anti-Tac (HAT) and Mik beta1 (HuMik beta 1) Abs directed at IL-2R alpha and IL-2R beta, respectively, inhibit IL-2 binding and biological activity and together act synergistically in vitro. The Abs have been used successfully in primate models of allograft rejection, graft-vs-host disease, and autoimmunity. We produced bifunctional humanized anti-IL-2R alpha beta Abs (BF-IgG) to combine the specificity of the two Abs into one entity by fusing HAT-producing NSO cells and HuMik beta 1-producing Sp2/0 cells. BF-IgG was purified using protein G-Sepharose affinity chromatography, followed by IL-2R alpha and IL-2R beta affinity chromatography and hydrophobic interaction chromatography. BF-IgG exhibited both anti-IL-2R alpha and anti-IL-2R beta specificities in binding assays. While the Ab binds the IL-2R with intermediate affinity (Kd = 2.82 nM), it does not inhibit IL-15 binding to its high affinity IL-15R. In Kit225/K6 (IL-2R alpha beta gamma+) cells, BF-IgG was 10-fold more potent than a HAT/HuMik beta 1 equimolar mixture in blocking IL-2-induced proliferation and, unexpectedly, was at least 65-fold more active than the mixture in blocking IL-15-induced proliferation. This dual inhibitory activity may be due to cross-linking of the IL-2R alpha and IL-2R beta, thus blocking IL-2 binding and possibly impeding the association of IL-2R beta with IL-15R. BF-IgG has potent immunosuppressant activities against both IL-2- and IL-15-mediated responses, and this antagonist could be more efficacious than HAT and/or HuMik beta 1 for the treatment of autoimmunity and the prevention of allograft rejection.


Subject(s)
Antibodies, Bispecific/pharmacology , Growth Inhibitors/immunology , Growth Inhibitors/pharmacology , Interleukin-15/physiology , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/physiology , Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/isolation & purification , Antibodies, Blocking/pharmacology , Antibody Affinity , Antibody Specificity , Binding Sites, Antibody , Cell Division/immunology , Clone Cells , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Hybrid Cells , Leukemia, T-Cell/immunology , Leukemia, T-Cell/metabolism , Sodium Dodecyl Sulfate , Tumor Cells, Cultured
12.
Eur J Immunol ; 27(5): 1152-63, 1997 May.
Article in English | MEDLINE | ID: mdl-9174605

ABSTRACT

Differentiation of thymocytes into mature single-positive T cells is an ordered process involving sequential interactions between T cell receptor (TCR), co-receptors (CD4 or CD8) and their appropriate major histocompatibility complex-encoded ligands. Precisely how these receptor/co-receptor engagements determine lineage commitment is still controversial, but recently it has been suggested that quantitative differences in the signal transmitted by co-ligation of CD4 versus CD8 with TCR might provide the discriminating signal. We examine this hypothesis, using bispecific F(ab')2 antibodies to mimic TCR/ co-receptor engagement during thymocyte differentiation. These bispecific antibodies lack Fc and can engage surface molecules without extensive cross-linking or targeting to Fc receptor-bearing cells. We show that TCR/CD3 co-ligation with CD4 induces efficient differentiation of mature CD4 lineage cells, irrespective of their TCR specificity. Interestingly, TCR/CD3 co-ligation with CD8 also induces maturation of CD4 T cells, although less efficiently, but not of CD8 T cells. Thus, although the signals delivered by co-ligation of TCR and CD8 appear weaker than from co-ligation of TCR and CD4, the outcome from either engagement is the same. These data suggest that differences in signal intensity alone do not determine lineage commitment in the thymus, but that distinct signals are required for CD4 and CD8 single-positive cell differentiation.


Subject(s)
CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology , Thymus Gland/immunology , Animals , Antibodies, Bispecific/pharmacology , CD3 Complex/immunology , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/classification , CD8 Antigens/immunology , Cell Differentiation/immunology , Crosses, Genetic , Immunoglobulin Fab Fragments/pharmacology , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Organ Culture Techniques , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell/immunology , Thymus Gland/cytology
13.
J Exp Med ; 185(8): 1413-22, 1997 Apr 21.
Article in English | MEDLINE | ID: mdl-9126922

ABSTRACT

Anti-CD3 monoclonal antibodies (mAbs) are potent immunosuppressive agents used in clinical transplantation. However, the activation-related adverse side effects associated with these mAbs have prompted the development of less toxic nonmitogenic anti-CD3 mAb therapies. At present, the functional and biochemical consequences of T cell exposure to nonmitogenic anti-CD3 is unclear. In this study, we have examined the early signaling events triggered by a nonmitogenic anti-CD3 mAb. Like the mitogenic anti-CD3 mAb, nonnmitogenic anti-CD3 triggered changes in the T cell receptor (TCR) complex, including zeta chain tyrosine phosphorylation and ZAP-70 association. However, unlike the mitogenic anti-CD3 stimulation, nonmitogenic anti-CD3 was ineffective at inducing the highly phosphorylated form of zeta (p23) and tyrosine phosphorylation of the associated ZAP-70 tyrosine kinase. This proximal signaling deficiency correlated with minimal phospholipase Cgamma-1 phosphorylation and failure to mobilize detectable Ca2+. Not only did biochemical signals delivered by nonmitogenic anti-CD3 resemble altered peptide ligand signaling, but exposure of Th1 clones to nonmitogenic anti-CD3 also resulted in functional anergy. Finally, a bispecific anti-CD3 X anti-CD4 F(ab)'2 reconstituted early signal transduction events and induced proliferation, suggesting that defective association of lck with the TCR complex may underlie the observed signaling differences between the mitogenic and nonmitogenic anti-CD3.


Subject(s)
CD3 Complex/immunology , Clonal Anergy , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , Immunosuppression Therapy/methods , Lymph Nodes/cytology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Phosphorylation , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor Aggregation , Signal Transduction , ZAP-70 Protein-Tyrosine Kinase
14.
J Immunol ; 157(8): 3420-9, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8871640

ABSTRACT

Peripheral T cell tolerance can be achieved through deletion of mature CD4+ cells activated by high dose Ag. We tested whether apoptosis of peripheral CD4+ cells could be induced by a stimulatory dose of Ag plus a soluble ligand to the nonpolymorphic epsilon-chain of the TCR-associated CD3 complex. CD4+ T cells from the DO10 mouse express a transgenic TCR-alphabeta specific for OVA peptide 323-339 presented by I-A(d). OVA alone induced clonal activation and expansion of peripheral CD4+/TCR transgene+ cells. Simultaneous exposure to specific Ag plus soluble anti-CD3 Fos, a nonmitogenic anti-CD3epsilon genetically engineered F(ab')2-like Ab, blocked expansion and induced death of CD4+/TCR transgene+ cells, but not CD4+/TCR transgene- T cells. In contrast, a mitogenic anti-CD3epsilon Ab induced polyclonal activation and nonselective T cell death. Sequential stimulation by Ag followed by anti-CD3 Fos also induced death of TCR transgene+ cells, whereas stimulation by anti-CD3 Fos followed by Ag did not affect cell viability or function. Anti-CD3 Fos-induced death was associated with DNA fragmentation characteristic of apoptosis, was facilitated by IL-2, and was initiated by stimulation during the S-G2 phases of the cell cycle. Anti-CD3 Fos could induce deletion of Ag-activated T cells by apoptosis in vivo. Thus, a soluble, non-Fc-binding anti-CD3 Ab can induce programmed cell death of Ag-activated peripheral CD4+ T cells by CD3epsilon cross-linking during S or G2. Peripheral T cell deletion by activation-driven apoptosis is under cell cycle control and can be exploited to achieve selective immunosuppression by nonmitogenic anti-CD3epsilon Abs.


Subject(s)
Apoptosis/immunology , CD3 Complex/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antigens/administration & dosage , Antigens/genetics , Cell Cycle , DNA Fragmentation , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/pharmacology , In Vitro Techniques , Interleukin-2/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Ovalbumin/administration & dosage , Ovalbumin/genetics , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Engineering , Proto-Oncogene Proteins c-fos/administration & dosage , Proto-Oncogene Proteins c-fos/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
15.
J Infect Dis ; 172(5): 1317-23, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7594670

ABSTRACT

Clostridium perfringens exotoxins have been implicated as major virulence factors responsible for shock and organ failure in gas gangrene, yet the mechanism(s) by which they mediate cardiovascular dysfunction remain enigmatic. Recombinant (r) phospholipase C (PLC), r theta-toxin, culture supernatant (crude toxin), or 0.9% NaCl was infused intravenously into awake rabbits. Cardiac index (CI), mean arterial pressure (MAP), heart rate (HR), central venous pressure (CVP), arterial blood gases, and hematocrit were measured 1 h before and for 3 h after toxin infusion. Crude toxin and rPLC decreased CI, MAP, and HR and increased CVP; mortality was 87.5% and 83%, respectively. r theta-toxin did not decrease CI or MAP and mortality was 25%. Further, crude toxin and rPLC but not r theta-toxin inhibited cardiac contractility (dF/dt) in isolated rabbit atrial muscles. These results suggest that PLC-induced myocardial dysfunction contributes to shock in C. perfringens infection.


Subject(s)
Bacterial Toxins/toxicity , Clostridium perfringens , Hemodynamics/drug effects , Hemolysin Proteins/toxicity , Type C Phospholipases/toxicity , Animals , Bacterial Toxins/administration & dosage , Blood Pressure/drug effects , Carbon Dioxide/blood , Central Venous Pressure/drug effects , Erythrocyte Volume/drug effects , Heart Rate/drug effects , Hemolysin Proteins/administration & dosage , Hemolysis , Hydrogen-Ion Concentration , Infusions, Intravenous , Male , Oxygen/blood , Partial Pressure , Rabbits , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Time Factors , Type C Phospholipases/administration & dosage , Vascular Resistance/drug effects
16.
J Hematother ; 4(5): 389-94, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8581374

ABSTRACT

The heterodimer-forming leucine zippers Fos and Jun can efficiently mediate the formation of bispecific F(ab')2 when they are fused separately to two different Fab' fragments. This recombinant method can be used in conjunction with the humanization process to yield humanized bispecific F(ab')2. The potential immunogenicity of the leucine zippers can be eliminated by their removal using pepsin digestion. This method has been scaled up to produce hundreds of milligrams of a bispecific F(ab')2 that targets two subunits of the human IL-2 receptor.


Subject(s)
Immunoglobulin Fab Fragments/immunology , Leucine Zippers , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Interleukin-2/immunology , Recombinant Fusion Proteins/immunology , Amino Acid Sequence , Antibody Specificity , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Leucine Zippers/drug effects , Leucine Zippers/genetics , Molecular Sequence Data , Oxidation-Reduction , Pepsin A/pharmacology , Protein Conformation , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
17.
J Immunol ; 155(3): 1544-55, 1995 Aug 01.
Article in English | MEDLINE | ID: mdl-7636216

ABSTRACT

145-2C11, a hamster mAb directed against the mouse CD3 complex, is a potent immunosuppressive agent. Upon initial treatment, 145-2C11 triggers a systemic release of multiple cytokines that is responsible for the acute toxicity of the mAb. This cellular activation is a consequence of the cross-linking between T lymphocytes and Fc gamma R-bearing cells, mediated by the high affinity of the hamster mAb for murine Fc gamma Rs. Repeated mAb injections result in the onset of a neutralizing humoral response. Therefore, there has been an increased interest in developing nonmitogenic forms of anti-CD3 mAbs, although it is not clear whether these Abs will retain immunosuppressive properties. To determine whether the initial cytokine production is necessary for the immunosuppressive properties and the immunogenicity of anti-CD3 mAbs in vivo, we have generated chimeric (hamster 145-2C11 F(ab')2 region/mouse Fc gamma portion) mAbs using murine isotypes with different affinities for Fc gamma Rs. The 145-2C11 and a chimeric IgG2a isotype, both of which bind murine Fc gamma Rs avidly, had similar activating, immunogenic, and immunosuppressive properties in mice. The administration of a chimeric IgG3 isotype with a very low affinity for murine Fc gamma Rs did not result in cytokine production, a humoral response against the mAb, or TCR desensitization. Nevertheless, prolongation of skin graft survival was similar in the IgG3, IgG2a, and 145-2C11-treated mice, indicating that Fc gamma R nonbinding anti-CD3 mAbs retain potent immunosuppressive properties in vivo while not being immunogenic. This enhanced therapeutic to toxic profile may be beneficial in clinical transplantation.


Subject(s)
CD3 Complex/immunology , Graft Enhancement, Immunologic , Graft Rejection/prevention & control , Immunosuppressive Agents/therapeutic use , Receptors, IgG/metabolism , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Base Sequence , Cloning, Molecular , Cricetinae , Cytokines/metabolism , Drug Evaluation, Preclinical , Genes, Immunoglobulin , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/toxicity , Isoantibodies/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Muromonab-CD3/immunology , Muromonab-CD3/toxicity , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/toxicity , Skin Transplantation/immunology , T-Lymphocytes, Cytotoxic/metabolism
18.
Int Immunol ; 7(6): 947-55, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7577803

ABSTRACT

In the present study we tested whether T cells retargeted with a bispecific antibody (bsAb) could block the growth of lung metastases of syngeneic mammary adenocarcinoma in immunocompetent mice. BALB/c mice were injected i.v. with tumor and i.p. with a genetically engineered bispecific F(ab')2 [bs(Fab')2] having specificity for murine CD3 epsilon chain and for the gp52 mouse mammary tumor viral glycoprotein, which is expressed on the tumor cells. The bs(Fab')2 was physically stable in blood and serum, was removed from the body with a half-time of 12-15 h, and accumulated in lymphoid tissue where it bound to T cells. We show that treatment of tumor bearing mice with the bs(Fab')2 significantly prolonged their survival relative to untreated controls. Two other genetically engineered bs(Fab')2s having specificity for murine CD3 epsilon chain and irrelevant antigens did not inhibit tumor growth. In addition, survival was not affected by bsAb therapy using a variant tumor cell line that expressed low levels of the gp52 target antigen. Inhibition of tumor growth was even more evident by histologic analysis. Treatment with the relevant bs(Fab')2 resulted in a marked reduction of tumor burden in lung sections taken on days 7, 9 and 11. This is the first report demonstrating that a bsAb can inhibit the growth of syngeneic solid tumor metastases in mice without addition of T cell activators.


Subject(s)
Adenocarcinoma/therapy , Antibodies, Bispecific/therapeutic use , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Experimental/therapy , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Animals , Antibodies, Bispecific/pharmacokinetics , Antigens, Neoplasm/immunology , Antigens, Viral, Tumor/immunology , CD3 Complex/immunology , Glycoproteins/immunology , Immunoglobulin Fab Fragments/therapeutic use , Lung Neoplasms/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/mortality , Mammary Tumor Virus, Mouse/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology
19.
Cancer Immunol Immunother ; 40(3): 182-90, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7728777

ABSTRACT

Bispecific antibodies with specificity for CD3 and a tumor antigen can redirect cytolytic T cells to kill tumor targets, regardless of their natural specificity. To assess the clinical potential of bispecific antibodies for treatment of human cancers we have, in the present study, adapted a totally synergeic mouse model to the targeting of mouse T cells against mouse tumors in immunocompetent mice. We show that gp52 of the mouse mammary tumor virus (MTV) can serve as a tumor-specific antigen for redirected cellular cytotoxicity. Chemically crosslinked and genetically engineered bispecific antibodies with specificities for gp52 and murine CD3 epsilon-chain induced activated mouse T cells to specifically lyse mouse mammary tumor cells from cultured lines and primary tumors from C3H-MTV+ mice. Retargeted T cells also blocked the growth of mammary tumors in vitro as well as their growth in syngeneic mice. These findings identify murine MTV-induced mammary adenocarcinomas as a solid-tumor, animal model for retargeting T cells with bispecific antibodies against syngeneic breast cancer.


Subject(s)
Adenocarcinoma/immunology , Antibodies, Bispecific/immunology , Cytotoxicity, Immunologic/immunology , Mammary Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Viral, Tumor/analysis , Antigens, Viral, Tumor/immunology , Cytotoxicity Tests, Immunologic , Mammary Tumor Virus, Mouse/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Receptor-CD3 Complex, Antigen, T-Cell/immunology
20.
J Immunol ; 152(5): 2385-92, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-8133049

ABSTRACT

Anti-CD3 x antitumor bispecific Ab can retarget T cell mediated lysis in an MHC-independent fashion and prevent tumor growth in animal models. Two bispecific Ab preparations that differ in the presence or absence of Fc were compared in the 38C13 immunocompetent murine lymphoma model to evaluate how functional Fc and T cell activation impact on response to bispecific Ab therapy. Bispecific (bs) IgG contained functional Fc and was purified from hybrid-hybridoma Ab product. Bsf(ab')2 lacked functional Fc, and was genetically constructed using the leucine zipper technique. In vitro, bsF(ab')2 induced tumor cell lysis by activated T cells more effectively than bsIgG. However, bsF(ab')2 failed to induce T cell activation in the absence of tumor cells, and did so more slowly than bsIgG when tumor cells were present. In vivo, bsIgG induced nonspecific T cell activation whereas bsF(ab')2 did not. In therapy experiments, bsIgG inhibited tumor growth in mice although a single dose of bsF(ab')2 had minimal antitumor effect. BsF(ab')2 was capable of preventing tumor growth and improving survival when mice were also treated with T cell activators (IL-2 or staphylococcal enterotoxin B), or given repeated bsF(ab')2 doses. We conclude that therapeutic response to bispecific Ab was not dependent on functional Fc, but did require T cell activation. The use of bifunctional constructs that lack functional Fc therefore allows for separate manipulation of T cell retargeting and T cell activation and deserves further evaluation as a potential immunotherapy for malignancy.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Neoplasm/therapeutic use , CD3 Complex , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/genetics , Cloning, Molecular , Cytotoxicity, Immunologic , DNA, Complementary/genetics , Immunoglobulin Fc Fragments , Lymphocyte Activation , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred C3H , Molecular Sequence Data , Tumor Cells, Cultured/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...