Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 50(7): 898-908, 2022 07.
Article in English | MEDLINE | ID: mdl-35545256

ABSTRACT

Fragment crystallizable (Fc) fusion is commonly used for extending the half-life of biotherapeutics such as cytokines. In this work, we studied the pharmacokinetics of Fc-fused interleukin-10 (IL-10) proteins that exhibited potent antitumor activity in mouse syngeneic tumor models. At pharmacologically active doses of ≥0.1 mg/kg, both mouse Fc-mouse IL-10 and human Fc-human IL-10, constructed as the C terminus of the Fc domain fused with IL-10 via a glycine-serine polypeptide linker, exhibited nonlinear pharmacokinetics after intravenous administration to mice at the doses of 0.05, 0.5, and 5 mg/kg. With a nominal dose ratio of 1:10:100; the ratio of the area under the curve for mouse Fc-mouse IL-10 and human Fc-human IL-10 was 1:181:1830 and 1:75:633, respectively. In contrast, recombinant mouse or human IL-10 proteins exhibited linear pharmacokinetics in mice. Compartmental analysis, using the Michaelis-Menten equation with the in vitro IL-10 receptor alpha binding affinity inputted as the Km, unified the pharmacokinetic data across the dose range. Additionally, nontarget-mediated clearance estimated for fusion proteins was ∼200-fold slower than that for cytokines, causing the manifestation of target-mediated drug disposition (TMDD) in the fusion protein pharmacokinetics. The experimental data generated with a mouse IL-10 receptor alpha-blocking antibody and a human Fc-human IL-10 mutant with a reduced receptor binding affinity showed significant improvements in pharmacokinetics, supporting TMDD as the cause of nonlinearity. Target expression and its effect on pharmacokinetics must be determined when considering using Fc as a half-life extension strategy, and pharmacokinetic evaluations need to be performed at a range of doses covering pharmacological activity. SIGNIFICANCE STATEMENT: Target-mediated drug disposition can manifest to affect the pharmacokinetics of a fragment crystallizable (Fc)-fused cytokine when the nontarget-mediated clearance of the cytokine is decreased due to neonatal Fc receptor-mediated recycling and molecular weight increases that reduce the renal clearance. The phenomenon was demonstrated with interleukin-10 Fc-fusion proteins in mice at pharmacologically active doses. Future drug designs using Fc as a half-life extension approach for cytokines need to consider target expression and its effect on pharmacokinetics at relevant doses.


Subject(s)
Interleukin-10 , Animals , Half-Life , Humans , Interleukin-10/pharmacokinetics , Mice , Receptors, Interleukin-10 , Recombinant Fusion Proteins/pharmacokinetics
2.
Cancer Immunol Res ; 7(10): 1633-1646, 2019 10.
Article in English | MEDLINE | ID: mdl-31431433

ABSTRACT

Elotuzumab (Elo) is an IgG1 monoclonal antibody targeting SLAMF7 (CS1, CRACC, and CD319), which is highly expressed on multiple myeloma (MM) cells, natural killer (NK) cells, and subsets of other leukocytes. By engaging with FcγRIIIA (CD16), Elo promotes potent NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) and macrophage-mediated antibody-dependent cellular phagocytosis (ADCP) toward SLAMF7+ MM tumor cells. Relapsed/refractory MM patients treated with the combination of Elo, lenalidomide, and dexamethasone have improved progression-free survival. We previously showed that Elo enhances NK cell activity via a costimulation mechanism, independent of CD16 binding. Here, we further studied the effect of Elo on cytotoxicity of CD16-negative NK-92 cells. Elo, but not other SLAMF7 antibodies, uniquely enhanced cytotoxicity mediated by CD16-negative NK-92 cells toward SLAMF7+ target cells. Furthermore, this CD16-independent enhancement of cytotoxicity required expression of SLAMF7 containing the full cytoplasmic domain in the NK cells, implicating costimulatory signaling. The CD16-independent costimulation by Elo was associated with increased expression of NKG2D, ICAM-1, and activated LFA-1 on NK cells, and enhanced cytotoxicity was partially reduced by NKG2D blocking antibodies. In addition, an Fc mutant form of Elo that cannot bind CD16 promoted cytotoxicity of SLAMF7+ target cells by NK cells from most healthy donors, especially if previously cultured in IL2. We conclude that in addition to promoting NK cell-mediated ADCC (CD16-dependent) responses, Elo promoted SLAMF7-SLAMF7 interactions in a CD16-independent manner to enhance NK cytotoxicity toward MM cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Multiple Myeloma/drug therapy , Signaling Lymphocytic Activation Molecule Family/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/immunology
3.
Sci Adv ; 5(2): eaav2437, 2019 02.
Article in English | MEDLINE | ID: mdl-30801016

ABSTRACT

While a fraction of cancer patients treated with anti-PD-1 show durable therapeutic responses, most remain unresponsive, highlighting the need to better understand and improve these therapies. Using an in vivo screening approach with a customized shRNA pooled library, we identified DDR2 as a leading target for the enhancement of response to anti-PD-1 immunotherapy. Using isogenic in vivo murine models across five different tumor histologies-bladder, breast, colon, sarcoma, and melanoma-we show that DDR2 depletion increases sensitivity to anti-PD-1 treatment compared to monotherapy. Combination treatment of tumor-bearing mice with anti-PD-1 and dasatinib, a tyrosine kinase inhibitor of DDR2, led to tumor load reduction. RNA-seq and CyTOF analysis revealed higher CD8+ T cell populations in tumors with DDR2 depletion and those treated with dasatinib when either was combined with anti-PD-1 treatment. Our work provides strong scientific rationale for targeting DDR2 in combination with PD-1 inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dasatinib/pharmacology , Discoidin Domain Receptor 2/antagonists & inhibitors , Drug Delivery Systems , Immunity, Cellular , Immunotherapy , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Discoidin Domain Receptor 2/immunology , Female , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Programmed Cell Death 1 Receptor/immunology
4.
Curr Top Microbiol Immunol ; 423: 13-34, 2019.
Article in English | MEDLINE | ID: mdl-30790079

ABSTRACT

Monoclonal antibodies can mediate antitumor activity by multiple mechanisms. They can bind directly to tumor receptors resulting in tumor cell death, or can bind to soluble growth factors, angiogenic factors, or their cognate receptors blocking signals required for tumor cell growth or survival. Monoclonal antibodies, upon binding to tumor cell, can also engage the host's immune system to mediate immune-mediated destruction of the tumor. The Fc portion of the antibody is essential in engaging the host immune system by fixing complement resulting in complement-mediated cytotoxicity (CDC) of the tumor, or by engaging Fc receptors for IgG (FcγR) expressed by leukocytes leading to antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP) of tumor cells. Antibodies whose Fc portion preferentially engage activating FcγRs have shown greater inhibition of tumor growth and metastasis. Monoclonal antibodies can also stimulate the immune system by binding to targets expressed on immune cells. These antibodies may stimulate antitumor immunity by antagonizing a negative regulatory signal, agonizing a costimulatory signal, or depleting immune cells that are inhibitory. The importance of Fc:FcγR interactions in antitumor therapy for each of these mechanisms have been demonstrated in both mouse models and clinical trials and will be the focus of this chapter.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Receptors, Fc/immunology , Receptors, IgG/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Humans , Neoplasms/pathology
5.
J Immunol ; 202(1): 151-159, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30530595

ABSTRACT

The FcγRs are immune cell surface proteins that bind IgG and facilitate cytokine production, phagocytosis, and Ab-dependent, cell-mediated cytotoxicity. FcγRs play a critical role in immunity; variation in these genes is implicated in autoimmunity and other diseases. Cynomolgus macaques are an excellent animal model for many human diseases, and Mauritian cynomolgus macaques (MCMs) are particularly useful because of their restricted genetic diversity. Previous studies of MCM immune gene diversity have focused on the MHC and killer cell Ig-like receptor. In this study, we characterize FcγR diversity in 48 MCMs using PacBio long-read sequencing to identify novel alleles of each of the four expressed MCM FcγR genes. We also developed a high-throughput FcγR genotyping assay, which we used to determine allele frequencies and identify FcγR haplotypes in more than 500 additional MCMs. We found three alleles for FcγR1A, seven each for FcγR2A and FcγR2B, and four for FcγR3A; these segregate into eight haplotypes. We also assessed whether different FcγR alleles confer different Ab-binding affinities by surface plasmon resonance and found minimal difference in binding affinities across alleles for a panel of wild type and Fc-engineered human IgG. This work suggests that although MCMs may not fully represent the diversity of FcγR responses in humans, they may offer highly reproducible results for mAb therapy and toxicity studies.


Subject(s)
Genotype , Macaca fascicularis , Receptors, IgG/genetics , Alleles , Animals , Antibody-Dependent Cell Cytotoxicity , Gene Frequency , Haplotypes , High-Throughput Nucleotide Sequencing , Humans , Immunity , Immunoglobulin G/metabolism , Models, Animal , Protein Binding/genetics , Receptors, IgG/metabolism
6.
MAbs ; 10(1): 95-103, 2018 01.
Article in English | MEDLINE | ID: mdl-29135326

ABSTRACT

TL1A, a tumor necrosis factor-like cytokine, is a ligand for the death domain receptor DR3. TL1A, upon binding to DR3, can stimulate lymphocytes and trigger secretion of proinflammatory cytokines. Therefore, blockade of TL1A/DR3 interaction may be a potential therapeutic strategy for autoimmune and inflammatory diseases. Recently, the anti-TL1A monoclonal antibody 1 (mAb1) with a strong potency in blocking the TL1A/DR3 interaction was identified. Here, we report on the use of hydrogen/deuterium exchange mass spectrometry (HDX-MS) to obtain molecular-level details of mAb1's binding epitope on TL1A. HDX coupled with electron-transfer dissociation MS provided residue-level epitope information. The HDX dataset, in combination with solvent accessible surface area (SASA) analysis and computational modeling, revealed a discontinuous epitope within the predicted interaction interface of TL1A and DR3. The epitope regions span a distance within the approximate size of the variable domains of mAb1's heavy and light chains, indicating it uses a unique mechanism of action to block the TL1A/DR3 interaction.


Subject(s)
Antibodies, Monoclonal/metabolism , Deuterium Exchange Measurement/methods , Epitope Mapping/methods , Epitopes/immunology , Mass Spectrometry/methods , Molecular Docking Simulation , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Binding Sites, Antibody , CHO Cells , Cricetulus , Humans , Kinetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Tumor Necrosis Factor, Member 25/immunology , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology
7.
Oncoimmunology ; 6(9): e1339853, 2017.
Article in English | MEDLINE | ID: mdl-28932638

ABSTRACT

Elotuzumab is a humanized therapeutic monoclonal antibody directed to the surface glycoprotein SLAMF7 (CS1, CRACC, CD319), which is highly expressed on multiple myeloma (MM) tumor cells. Improved clinical outcomes have been observed following treatment of MM patients with elotuzumab in combination with lenalidomide or bortezomib. Previous work showed that elotuzumab stimulates NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC), via Fc-domain engagement with FcγRIIIa (CD16). SLAMF7 is also expressed on NK cells, where it can transmit stimulatory signals. We tested whether elotuzumab can directly activate NK cells via ligation with SLAMF7 on NK cells in addition to targeting ADCC through CD16. We show that elotuzumab strongly promoted degranulation and activation of NK cells in a CD16-dependent manner, and a non-fucosylated form of elotuzumab with higher affinity to CD16 exhibited enhanced potency. Using F(ab')2 or Fc-mutant forms of the antibody, the direct binding of elotuzumab to SLAMF7 alone could not stimulate measurable CD69 expression or degranulation of NK cells. However, the addition of soluble elotuzumab could costimulate calcium signaling responses triggered by multimeric engagement of NKp46 and NKG2D in a CD16-independent manner. Thus, while elotuzumab primarily stimulates NK cells through CD16, it can also transduce effective "trans"-costimulatory signals upon direct engagement with SLAMF7, since these responses did not require direct co-engagement with the activating receptors. Trans-costimulation by elotuzumab has potential to reduce activation thresholds of other NK cell receptors engaging with their ligands on myeloma target cell surfaces, thereby potentially further increasing NK cell responsiveness in patients.

8.
Cancer Immunol Res ; 5(2): 106-117, 2017 02.
Article in English | MEDLINE | ID: mdl-28073774

ABSTRACT

Antibody blockade of programmed death-1 (PD-1) or its ligand, PD-L1, has led to unprecedented therapeutic responses in certain tumor-bearing individuals, but PD-L1 expression's prognostic value in stratifying cancer patients for such treatment remains unclear. Reports conflict on the significance of correlations between PD-L1 on tumor cells and positive clinical outcomes to PD-1/PD-L1 blockade. We investigated this issue using genomically related, clonal subsets from the same methylcholanthrene-induced sarcoma: a highly immunogenic subset that is spontaneously eliminated in vivo by adaptive immunity and a less immunogenic subset that forms tumors in immunocompetent mice, but is sensitive to PD-1/PD-L1 blockade therapy. Using CRISPR/Cas9-induced loss-of-function approaches and overexpression gain-of-function techniques, we confirmed that PD-L1 on tumor cells is key to promoting tumor escape. In addition, the capacity of PD-L1 to suppress antitumor responses was inversely proportional to tumor cell antigenicity. PD-L1 expression on host cells, particularly tumor-associated macrophages (TAM), was also important for tumor immune escape. We demonstrated that induction of PD-L1 on tumor cells was IFNγ-dependent and transient, but PD-L1 induction on TAMs was of greater magnitude, only partially IFNγ dependent, and was stable over time. Thus, PD-L1 expression on either tumor cells or host immune cells could lead to tumor escape from immune control, indicating that total PD-L1 expression in the immediate tumor microenvironment may represent a more accurate biomarker for predicting response to PD-1/PD-L1 blockade therapy, compared with monitoring PD-L1 expression on tumor cells alone. Cancer Immunol Res; 5(2); 106-17. ©2017 AACR.


Subject(s)
B7-H1 Antigen/genetics , Gene Expression , Neoplasms/genetics , Neoplasms/immunology , Tumor Escape/genetics , Tumor Escape/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Gene Knockout Techniques , Genes, MHC Class I/genetics , Genes, MHC Class I/immunology , Humans , Male , Mice , Mutation , Neoplasms/pathology , Sarcoma/genetics , Sarcoma/immunology , Sarcoma/pathology , Tumor Burden , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
Blood Adv ; 1(12): 753-765, 2017 May 09.
Article in English | MEDLINE | ID: mdl-29296719

ABSTRACT

Elotuzumab, a humanized monoclonal antibody that binds human signaling lymphocytic activation molecule F7 (hSLAMF7) on myeloma cells, was developed to treat patients with multiple myeloma (MM). Elotuzumab has a dual mechanism of action that includes the direct activation of natural killer (NK) cells and the induction of NK cell-mediated antibody-dependent cellular cytotoxicity. This study aimed to characterize the effects of elotuzumab on NK cells in vitro and in patients with MM and to determine whether elotuzumab antitumor activity was improved by programmed death receptor-1 (PD-1) blockade. Elotuzumab promoted NK cell activation when added to a coculture of human NK cells and SLAMF7-expressing myeloma cells. An increased frequency of activated NK cells was observed in bone marrow aspirates from elotuzumab-treated patients. In mouse tumor models expressing hSLAMF7, maximal antitumor efficacy of a murine immunoglobulin G2a version of elotuzumab (elotuzumab-g2a) required both Fcγ receptor-expressing NK cells and CD8+ T cells and was significantly enhanced by coadministration of anti-PD-1 antibody. In these mouse models, elotuzumab-g2a and anti-PD-1 combination treatment promoted tumor-infiltrating NK and CD8+ T-cell activation, as well as increased intratumoral cytokine and chemokine release. These observations support the rationale for clinical investigation of elotuzumab/anti-PD-1 combination therapy in patients with MM.

10.
Clin Cancer Res ; 15(10): 3376-83, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19401346

ABSTRACT

PURPOSE: This study was undertaken to evaluate the effects of MDX-1401, a nonfucosylated fully human monoclonal antibody that binds to human CD30, and to determine whether it exhibits greater in vitro and in vivo activity than its parental antibody. EXPERIMENTAL DESIGN: Assays measuring antibody binding to CD30-expressing cells and FcgammaRIIIa (CD16) transfectants as well as antibody-dependent cellular cytotoxicity (ADCC) were conducted. Antitumor activity was determined using a Karpas-299 systemic model. RESULTS: The binding of MDX-1401 to CD30 antigen was identical to fucose-containing parental anti-CD30 antibody (MDX-060). In contrast, MDX-1401 showed increased binding affinity to FcgammaRIIIa-transfected cells resulting in increased effector function. MDX-1401 greatly improved ADCC activity as evidenced by a decrease in half-maximal effective concentration (EC(50)) and an increase in maximum cell lysis when compared with MDX-060. Increased ADCC activity was observed among a panel of cell lines, including one with very low CD30 antigen expression in which parental antibody failed to induce any detectable ADCC. MDX-1401 activity with all FcgammaRIIIa polymorphic variants, including less active Phe/Phe158 and Phe/Val158 effector cells, was shown. Furthermore, MDX-1401 was efficacious in inhibiting tumor growth in CD30(+) lymphoma xenografts. CONCLUSIONS: The low doses of antibody required for ADCC activity irrespective of donor genotype, the ability to mediate ADCC in target cells expressing low levels of CD30, and increased in vivo efficacy support the development of MDX-1401 for treatment of malignant lymphoma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Lymphoma/drug therapy , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibody Affinity/drug effects , Antibody Affinity/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Binding Sites, Antibody/immunology , CHO Cells , Carbohydrates/chemistry , Carbohydrates/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fucose/chemistry , Fucose/immunology , Humans , Ki-1 Antigen/immunology , Lymphoma/immunology , Lymphoma/pathology , Male , Mice , Mice, SCID , Receptors, IgG/chemistry , Receptors, IgG/immunology
11.
Br J Haematol ; 132(3): 317-25, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16409296

ABSTRACT

Expression of the type I receptor for Fc domain of immunoglobulin (Ig)G (Fc gammaRI or CD64) is restricted to myeloid effector cells, such as monocytes, macrophages and a subset of dendritic cells. Previous work has indicated a role for Fc gammaRI in antibody-dependent phagocytosis and lysis of tumour cells. We hypothesised that tagging of tumour cells with an anti-Fc gammaRI single chain Fv (sFv) may facilitate targeting to this receptor on effector cells, thereby initiating tumour cytotoxicity. A vector encoding the sFv for an Fc gammaRI-specific antibody (H22), linked to the transmembrane domain of platelet-derived growth factor was constructed. Transfected tumour cells expressed high surface levels of functional H22-sFv, which greatly enhanced susceptibility for phagocytosis and lysis by monocytes and macrophages. The expression of H22-sFv evoked the ability of tumour cells to directly activate monocytes, as evidenced by phosphorylation of mitogen-activated protein kinase and secretion of the inflammatory cytokines interleukin (IL)-1beta, tumour necrosis factor-alpha and IL-6. Moreover, growth of tumour cells in mice expressing H22-sFv was profoundly delayed (or absent) in transgenic mice expressing human Fc gammaRI. These results demonstrated that tumour cells can be readily modified to activate cell effector mechanisms, a strategy that may be useful for in vivo targeting in patients.


Subject(s)
Neoplasms/genetics , Receptors, IgG/genetics , Animals , Antibodies, Neoplasm/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Genetic Vectors/genetics , Humans , Interleukin-1/immunology , Interleukin-6/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Neoplasms/immunology , Phagocytosis/genetics , Phagocytosis/immunology , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, IgG/immunology , Transfection , Tumor Necrosis Factor-alpha/immunology
12.
Immunol Invest ; 34(4): 417-29, 2005.
Article in English | MEDLINE | ID: mdl-16304730

ABSTRACT

There is a continuing need for alternatives to current human adjuvants. Recombinant protein vaccines, which target antigen to human Fc gamma receptor type I (hFcgammaRI) on hFcgammaRI-expressing antigen presenting cells, provide one potential alternative. Using a recombinant anti-hFcgammaRI-antigen fusion protein and adjuvant independent mouse model, we demonstrate enhanced antigen-specific antibody responses to low doses of antigen, when targeting antigen to hFcgammaRI in vivo. Enhanced antibody production to hFcyRI-targeted antigen is evident in both primary and secondary immune responses, as compared to that of non-targeted antigen. Furthermore, antibody isotype and cytokine responses following immunization with hFcgammaRI-targeted antigen, suggest enhancement of both Th1 and Th2 responses.


Subject(s)
Adjuvants, Immunologic/genetics , Antibody Formation , Antigens/immunology , Cytokines/biosynthesis , Immunoglobulin Variable Region/immunology , Receptors, IgG/immunology , Animals , Humans , Immunization , Immunoglobulin Variable Region/genetics , Male , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/immunology , Streptavidin/genetics , Streptavidin/immunology
13.
Curr Opin Mol Ther ; 7(2): 157-63, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15844624

ABSTRACT

The use of monoclonal antibodies for immunotherapy has been validated by the commercialization of multiple monoclonal antibody products for oncology, infectious diseases and autoimmune diseases. In addition to their application as 'naked' antibodies, they have been used as delivery vehicles for cytotoxic agents to cancer cells. The exquisite specificity of antibodies can also be exploited to initiate and/or enhance the immune response to tumors or infectious agents by targeting the relevant antigen to antigen-presenting cells (APCs). Such antibody-targeted vaccines (ATVs) have demonstrated remarkable activity in preclinical models by eliminating the need for adjuvant and repetitive boosting, overcoming immunological non-responsiveness, inducing mucosal immunity and eliciting therapeutic cytotoxic T-cell-mediated antitumor effects. A variety of different receptors on APCs have been exploited for targeting antigens, which may allow optimization for specific immune responses. Selective targeting to appropriate receptors on APC subsets combined with strong activating signals is important for generating potent cytolytic T-cell responses. Studies have also suggested that ATVs may be selectively exploited to induce tolerance against antigens for treatment against transplant rejection or autoimmune diseases. The clinical development of this new class of antibody-based products has recently been initiated.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/drug effects , Vaccines/pharmacology , Animals , Antibodies, Monoclonal/genetics , Antigen-Presenting Cells/immunology , Humans , Vaccines/immunology
14.
J Infect Dis ; 191(4): 507-14, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15655773

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome (SARS) remains a significant public health concern after the epidemic in 2003. Human monoclonal antibodies (MAbs) that neutralize SARS-associated coronavirus (SARS-CoV) could provide protection for exposed individuals. METHODS: Transgenic mice with human immunoglobulin genes were immunized with the recombinant major surface (S) glycoprotein ectodomain of SARS-CoV. Epitopes of 2 neutralizing MAbs derived from these mice were mapped and evaluated in a murine model of SARS-CoV infection. RESULTS: Both MAbs bound to S glycoprotein expressed on transfected cells but differed in their ability to block binding of S glycoprotein to Vero E6 cells. Immunoprecipitation analysis revealed 2 antibody-binding epitopes: one MAb (201) bound within the receptor-binding domain at aa 490-510, and the other MAb (68) bound externally to the domain at aa 130-150. Mice that received 40 mg/kg of either MAb prior to challenge with SARS-CoV were completely protected from virus replication in the lungs, and doses as low as 1.6 mg/kg offered significant protection. CONCLUSIONS: Two neutralizing epitopes were defined for MAbs to SARS-CoV S glycoprotein. Antibodies to both epitopes protected mice against SARS-CoV challenge. Clinical trials are planned to test MAb 201, a fully human MAb specific for the epitope within the receptor-binding region.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Immunization, Passive , Membrane Glycoproteins/immunology , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins/immunology , Animals , Cells, Cultured , Disease Models, Animal , Epitope Mapping , Epitopes/immunology , Female , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutralization Tests , Protein Binding , Spike Glycoprotein, Coronavirus
15.
Methods Mol Biol ; 283: 71-85, 2004.
Article in English | MEDLINE | ID: mdl-15197303

ABSTRACT

This chapter discusses two related methods for creating Fab' x Fab' chemically linked BsAb. Both methods require the generation of purified F(ab')2 fragments of each antibody and use reagents that react with the free thiols generated upon reduction of interheavy chain disulfide bonds of the F(ab')2 fragments. Upon reduction, the resulting Fabs are then recombined to form a Fab' x Fab' BsAb. 5,5'-dithiobis (2-nitrobenzoic acid) (DTNB) acts to regenerate disulfide bonds between the two Fabs, whereas o-phenylenedimaleimide (o-PDM) acts to form a thioether bond between the two Fabs. After coupling, the bispecific antibody is purified from the uncoupled Fabs by size-exclusion chromatography. The advantages and disadvantages of each conjugation method are discussed.


Subject(s)
Antibodies, Bispecific/chemistry , Immunoglobulin Fragments/chemistry , Chromatography, Gel , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel
16.
J Immunol ; 171(11): 6251-9, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634142

ABSTRACT

The immune modulatory molecule CTLA-4 (CD152), through interactions with the B7 costimulatory molecules, has been shown to be a negative regulator of T cell activation in various murine model systems. Abs that block CTLA-4 function can enhance immune responses that mediate potent antitumor activity. However, CTLA-4 blockade can also exacerbate autoimmune disease. The safety and activity of anti-CTLA-4 Abs in primates has not been addressed. To that end, we generated human Abs against CTLA-4 using transgenic mice expressing human Ig genes. A high affinity Ab (10D1) that blocked the binding of CTLA-4 to the B7-1 and B7-2 ligands and had cross-reactivity with macaque CTLA-4 was chosen for further development. Administration of 10D1 to cynomolgus macaques significantly enhanced Ab responses to hepatitis surface Ag and a human melanoma cell vaccine. Anti-self Ab responses as measured by immunoassays using lysate from melanocyte-rich tissues were elicited in those animals receiving the melanoma cell vaccine and anti-CTLA-4 Ab. Remarkably, chronic administration of 10D1 did not result in measurable polyclonal T cell activation, significant alteration of the lymphocyte subsets, or induce clinically observable autoimmunity. Repeated dosing of the 10D1 did not elicit monkey anti-human Ab responses in the monkeys. These observations support the development of CTLA-4 blockade for human immunotherapy.


Subject(s)
Antibodies, Blocking/adverse effects , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/adverse effects , Antigens, Differentiation/immunology , Cancer Vaccines/pharmacology , Down-Regulation/immunology , Hepatitis B Vaccines/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Blocking/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/biosynthesis , Antigens, CD , CTLA-4 Antigen , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Drug Synergism , Female , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/immunology , Humans , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/immunology , Macaca fascicularis , Male , Mice , Mice, Transgenic
17.
Blood ; 102(10): 3737-42, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12881320

ABSTRACT

CD30 is a promising target for antibody-based immunotherapy of Hodgkin lymphoma (HL) and anaplastic large cell lymphoma. To overcome the limitations from currently available murine anti-CD30 monoclonal antibodies (mAbs), a new fully human anti-CD30 antibody was generated. Binding properties were evaluated by recombinant CD30 capture enzyme-linked immunosorbent assay (ELISA) and fluorescence-activated cell-sorter (FACS) flow cytometry. Activity of this new mAb was assessed in vitro using growth inhibition and antibody-dependent cellular cytotoxicity (ADCC) assays on several cell lines. In vivo activity was determined in a solid as well as in a disseminated xenografted model of HL in severe combined immunodeficiency (SCID) mice. The mAb 5F11 showed specific binding to CD30 (cluster A). The ADCC assays indicated dose-dependent lysis of L540 cells when 5F11 was combined with human effector cells. Upon cross-linking in vitro, 5F11 inhibited the growth of CD30-expressing cell lines. In vivo, treatment with 5F11 induced a marked growth delay or even a complete regression of established xenografted HL in SCID mice. In the disseminated HL model, a high proportion of 5F11-treated mice experienced long-term survival. The new human anti-CD30 monoclonal antibody 5F11 shows promise as a means of CD30-targeted immunotherapy of malignant lymphomas. Based on these results, a clinical phase 1 study in patients with refractory CD30+ lymphoma has been initiated.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Ki-1 Antigen/immunology , Lymphoma/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antigens, Neoplasm/immunology , Cell Division , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Hodgkin Disease/drug therapy , Hodgkin Disease/mortality , Humans , Lymphoma/mortality , Mice , Mice, SCID , Neoplasm Transplantation , Survival Rate , Transplantation, Heterologous
18.
Biomol Eng ; 20(1): 21-33, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12485681

ABSTRACT

The professional antigen presenting cell (APC) plays an essential role in the initiation and propagation of the acquired immune response. Thus, much work has been done in designing strategies that target vaccine antigen (Ag) to APC. Utilizing recombinant DNA technology, we have created a unique two-component system that delivers biotinylated Ag to the Fc gamma receptor type I (FcgammaRI) on APC. Our studies demonstrate that we can successfully engineer FcgammaRI-specific targeting element proteins that simultaneously bind both biotin and recognize FcgammaRI. Additionally, we are able to engineer biotinylated Ag, which form functional elements when adsorbed onto latex microspheres. Furthermore, the targeting and functional element components bind to each other and successfully form two-component immunogens. T-cell activation in response to targeted Ag-laden microspheres is 10- to 100-fold greater than the response to the non-targeted Ag-laden microspheres. This enhancement is 100- to 1000-fold greater than the responses generated to soluble Ag. Thus, our results suggest that specific targeting of Ag-laden microspheres to FcgammaRI may significantly enhance the adjuvant properties of microparticulate delivery systems. Further development of this system may help to elucidate the mechanisms involved in generating enhanced responses to APC-targeted vaccines and significantly advance vaccine technology.


Subject(s)
Antigen-Presenting Cells/immunology , Drug Delivery Systems/methods , Lymphocyte Activation/drug effects , Receptors, IgG/administration & dosage , Streptavidin/administration & dosage , T-Lymphocytes/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Antigens , Biotinylation/methods , Cell Communication/physiology , Cells, Cultured , Drug Combinations , Humans , Microspheres , Quality Control , Receptors, IgG/immunology , Receptors, IgG/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Streptavidin/immunology , Streptavidin/metabolism , Structure-Activity Relationship , T-Lymphocytes/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...