Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38987489

ABSTRACT

PURPOSE: Immune cells are capable of eliminating leukemic cells, as evidenced by outcomes in hematopoietic cell transplantation (HCT). However, patients who fail induction therapy will not benefit from HCT due to their minimal residual disease (MRD) status. Thus, we aimed to develop an immunomodulatory agent to reduce MRD by activating immune effector cells in the presence of leukaemia cells via a novel fusion protein that chimerises two clinically tolerated biologics: a CD33 antibody and the IL15Ra/IL15 complex (CD33xIL15). METHODS: We generated a set of CD33xIL15 fusion protein constructs with varying configurations and identified those with the best in vitro AML-binding, T cell activation, and NK cell potentiation. Using 89Zr-immunoPET imaging we then evaluated the biodistribution and in vivo tumour retention of the most favourable CD33xIL15 constructs in an AML xenograft model. Ex vivo biodistribution studies were used to confirm the pharmacokinetics of the constructs. RESULTS: Two of the generated fusion proteins, CD33xIL15 (N72D) and CD33xIL15wt, demonstrated optimal in vitro behaviour and were further evaluated in vivo. These studies revealed that the CD33xIL15wt candidate was capable of being retained in the tumour for as long as its parental CD33 antibody, Lintuzumab (13.9 ± 3.1%ID/g vs 18.6 ± 1.1%ID/g at 120 h). CONCLUSION: This work demonstrates that CD33xIL15 fusion proteins are capable of targeting leukemic cells and stimulating local T cells in vitro and of concentrating in the tumour in AML xenografts. It also highlights the importance of 89Zr-immunoPET to guide the development and selection of tumour-targeted antibody-cytokine fusion proteins.

2.
Nature ; 611(7937): 787-793, 2022 11.
Article in English | MEDLINE | ID: mdl-36323781

ABSTRACT

Emerging studies indicate that cooperation between neurons and immune cells regulates antimicrobial immunity, inflammation and tissue homeostasis. For example, a neuronal rheostat provides excitatory or inhibitory signals that control the functions of tissue-resident group 2 innate lymphoid cells (ILC2s) at mucosal barrier surfaces1-4. ILC2s express NMUR1, a receptor for neuromedin U (NMU), which is a prominent cholinergic neuropeptide that promotes ILC2 responses5-7. However, many functions of ILC2s are shared with adaptive lymphocytes, including the production of type 2 cytokines8,9 and the release of tissue-protective amphiregulin (AREG)10-12. Consequently, there is controversy regarding whether innate lymphoid cells and adaptive lymphocytes perform redundant or non-redundant functions13-15. Here we generate a new genetic tool to target ILC2s for depletion or gene deletion in the presence of an intact adaptive immune system. Transgenic expression of iCre recombinase under the control of the mouse Nmur1 promoter enabled ILC2-specific deletion of AREG. This revealed that ILC2-derived AREG promotes non-redundant functions in the context of antiparasite immunity and tissue protection following intestinal damage and inflammation. Notably, NMU expression levels increased in inflamed intestinal tissues from both mice and humans, and NMU induced AREG production in mouse and human ILC2s. These results indicate that neuropeptide-mediated regulation of non-redundant functions of ILC2s is an evolutionarily conserved mechanism that integrates immunity and tissue protection.


Subject(s)
Immunity, Innate , Intestinal Mucosa , Lymphocytes , Neuropeptides , Animals , Humans , Mice , Cytokines/immunology , Cytokines/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/parasitology , Inflammation/pathology , Lymphocytes/immunology , Neuropeptides/metabolism , Neuropeptides/physiology , Amphiregulin , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology
3.
JCI Insight ; 7(5)2022 03 08.
Article in English | MEDLINE | ID: mdl-35260532

ABSTRACT

Phosphopeptides derived from dysregulated protein phosphorylation in cancer cells can be processed and presented by MHC class I and class II molecules and, therefore, represent an untapped class of tumor-specific antigens that could be used as widely expressed "public" cancer neoantigens (NeoAgs). We generated a TCR mimic (TCRm) mAb, 6B1, specific for a phosphopeptide derived from insulin receptor substrate 2 (pIRS2) presented by HLA-A*02:01. The pIRS2 epitope's presentation by HLA-A*02:01 was confirmed by mass spectrometry. The TCRm 6B1 specifically bound to pIRS2/HLA-A2 complex on tumor cell lines that expressed pIRS2 in the context of HLA-A*02:01. Bispecific mAbs engaging CD3 of T cells were able to kill tumor cell lines in a pIRS2- and HLA-A*02:01-restricted manner. Structure modeling shows a prerequisite for an arginine or lysine at the first position to bind mAb. Therefore, 6B1 could recognize phosphopeptides derived from various phosphorylated proteins with similar amino acid compositions. This raised the possibility that a TCRm specific for the pIRS2/HLA-A2 complex could target a range of phosphopeptides presented by HLA-A*02:01 in various tumor cells. This is the first TCRm mAb to our knowledge targeting a phosphopeptide/MHC class I complex; the potential of this class of agents for clinical applications warrants further investigation.


Subject(s)
HLA-A2 Antigen , Phosphopeptides , Antibodies, Monoclonal/metabolism , Insulin Receptor Substrate Proteins/metabolism , Phosphopeptides/metabolism , Receptors, Antigen, T-Cell/metabolism
4.
Sci Transl Med ; 14(636): eabe8195, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35294260

ABSTRACT

Most patients with non-small cell lung cancer (NSCLC) do not achieve durable clinical responses from immune checkpoint inhibitors, suggesting the existence of additional resistance mechanisms. Nicotinamide adenine dinucleotide (NAD)-induced cell death (NICD) of P2X7 receptor (P2X7R)-expressing T cells regulates immune homeostasis in inflamed tissues. This process is mediated by mono-adenosine 5'-diphosphate (ADP)-ribosyltransferases (ARTs). We found an association between membranous expression of ART1 on tumor cells and reduced CD8 T cell infiltration. Specifically, we observed a reduction in the P2X7R+ CD8 T cell subset in human lung adenocarcinomas. In vitro, P2X7R+ CD8 T cells were susceptible to ART1-mediated ADP-ribosylation and NICD, which was exacerbated upon blockade of the NAD+-degrading ADP-ribosyl cyclase CD38. Last, in murine NSCLC and melanoma models, we demonstrate that genetic and antibody-mediated ART1 inhibition slowed tumor growth in a CD8 T cell-dependent manner. This was associated with increased infiltration of activated P2X7R+CD8 T cells into tumors. In conclusion, we describe ART1-mediated NICD as a mechanism of immune resistance in NSCLC and provide preclinical evidence that antibody-mediated targeting of ART1 can improve tumor control, supporting pursuit of this approach in clinical studies.


Subject(s)
ADP Ribose Transferases , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , T-Lymphocyte Subsets , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , Adenosine Diphosphate , Animals , Carcinoma, Non-Small-Cell Lung/immunology , GPI-Linked Proteins/genetics , Humans , Lung Neoplasms/immunology , Mice
5.
Proc Natl Acad Sci U S A ; 117(14): 7981-7989, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32209664

ABSTRACT

Human infection by Zika virus (ZIKV) during pregnancy can lead to vertical transmission and fetal aberrations, including microcephaly. Prophylactic administration of antibodies can diminish or prevent ZIKV infection in animal models, but whether passive immunization can protect nonhuman primates and their fetuses during pregnancy has not been determined. Z004 and Z021 are neutralizing monoclonal antibodies to domain III of the envelope (EDIII) of ZIKV. Together the two antibodies protect nonpregnant macaques against infection even after Fc modifications to prevent antibody-dependent enhancement (ADE) in vitro and extend their half-lives. Here we report on prophylactic coadministration of the Fc-modified antibodies to pregnant rhesus macaques challenged three times with ZIKV during first and second trimester. The two antibodies did not entirely eliminate maternal viremia but limited vertical transmission, protecting the fetus from neurologic damage. Thus, maternal passive immunization with two antibodies to EDIII can shield primate fetuses from the harmful effects of ZIKV.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy Complications, Infectious/prevention & control , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Animals, Newborn , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Disease Models, Animal , Drug Therapy, Combination , Female , Fetus/immunology , Fetus/virology , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/administration & dosage , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Protein Engineering , RNA, Viral/isolation & purification , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/transmission , Zika Virus Infection/virology
6.
MAbs ; 7(5): 931-45, 2015.
Article in English | MEDLINE | ID: mdl-26073904

ABSTRACT

Bi-specific antibodies (BsAbs), which can simultaneously block 2 tumor targets, have emerged as promising therapeutic alternatives to combinations of individual monoclonal antibodies. Here, we describe the engineering and development of a novel, human bi-functional antibody-receptor domain fusion molecule with ligand capture (bi-AbCap) through the fusion of the domain 2 of human vascular endothelial growth factor receptor 1 (VEGFR1) to an antibody directed against insulin-like growth factor - type I receptor (IGF-IR). The bi-AbCap possesses excellent stability and developability, and is the result of minimal engineering. Beyond potent neutralizing activities against IGF-IR and VEGF, the bi-AbCap is capable of cross-linking VEGF to IGF-IR, leading to co-internalization and degradation of both targets by tumor cells. In multiple mouse xenograft tumor models, the bi-AbCap improves anti-tumor activity over individual monotherapies. More importantly, it exhibits superior inhibition of tumor growth, compared with the combination of anti-IGF-IR and anti-VEGF therapies, via powerful blockade of both direct tumor cell growth and tumor angiogenesis. The unique "capture-for-degradation" mechanism of the bi-AbCap is informative for the design of next-generation bi-functional anti-cancer therapies directed against independent signaling pathways. The bi-AbCap design represents an alternative approach to the creation of dual-targeting antibody fusion molecules by taking advantage of natural receptor-ligand interactions.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Neutralizing/pharmacology , Receptors, Somatomedin/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Nude , Microscopy, Confocal , Neoplasms, Experimental/drug therapy , Protein Stability , Receptor, IGF Type 1 , Surface Plasmon Resonance , Xenograft Model Antitumor Assays
7.
Cancer Biol Ther ; 15(9): 1208-18, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24921944

ABSTRACT

Stem cell factor receptor (c-Kit) exerts multiple biological effects on target cells upon binding its ligand stem cell factor (SCF). Aberrant activation of c-Kit results in dysregulated signaling and is implicated in the pathogenesis of numerous cancers. The development of more specific and effective c-Kit therapies is warranted given its essential role in tumorigenesis. In this study, we describe the biological properties of CK6, a fully human IgG1 monoclonal antibody against the extracellular region of human c-Kit. CK6 specifically binds c-Kit receptor with high affinity (EC 50 = 0.06 nM) and strongly blocks its interaction with SCF (IC 50 = 0.41 nM) in solid phase assays. Flow cytometry shows CK6 binding to c-Kit on the cell surface of human small cell lung carcinoma (SCLC), melanoma, and leukemia tumor cell lines. Furthermore, exposure to CK6 inhibits SCF stimulation of c-Kit tyrosine kinase activity and downstream signaling pathways such as mitogen-activated protein kinase (MAPK) and protein kinase B (AKT), in addition to reducing tumor cell line growth in vitro. CK6 treatment significantly decreases human xenograft tumor growth in NCI-H526 SCLC (T/C% = 57) and Malme-3M melanoma (T/C% = 58) models in vivo. The combination of CK6 with standard of care chemotherapy agents, cisplatin and etoposide for SCLC or dacarbazine for melanoma, more potently reduces tumor growth (SCLC T/C% = 24, melanoma T/C% = 38) compared with CK6 or chemotherapy alone. In summary, our results demonstrate that CK6 is a c-Kit antagonist antibody with tumor growth neutralizing properties and are highly suggestive of potential therapeutic application in treating human malignancies harboring c-Kit receptor.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoglobulin G/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Proto-Oncogene Proteins c-kit/metabolism , Small Cell Lung Carcinoma/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Etoposide/administration & dosage , Female , Heterografts , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Signal Transduction , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
8.
Biotechnol Bioeng ; 110(11): 2928-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23740533

ABSTRACT

Recent advances in mammalian cell culture processes have significantly increased product titers, but have also resulted in substantial increases in cell density and cellular debris as well as process and product related impurities. As such, with improvements in titer, corresponding improvements in downstream processing are essential. In this study we have developed an alternative antibody harvest process that incorporates flocculation using a novel stimulus responsive polymer, benzylated poly(allylamine), followed by depth filtration. As tested on multiple antibodies, this process demonstrates high process yield, improved clearance of cells and cell debris, and efficient reduction of aggregates, host cell proteins (HCP) and DNA. A wide operating window was established for this novel flocculation process through design of experiments condition screening and optimization. Residual levels of impurities in the Protein A eluate were achieved that potentially meet requirements of drug substance and thus alleviate the burden for further impurities removal in subsequent chromatography steps. In addition, efficient clearance of residual polymer was demonstrated using a fluorescence tagged polymer in the presence of a stimulus reagent. The mechanism of HCP and aggregates removal during flocculation was also explored. This novel and efficient process can be easily integrated into current mAb purification platforms, and may overcome downstream processing challenges.


Subject(s)
Antibodies/isolation & purification , Biological Products/isolation & purification , Chemical Fractionation/methods , Filtration/methods , Flocculation , Technology, Pharmaceutical/methods , Animals , CHO Cells , Cell Culture Techniques/methods , Cricetinae , Cricetulus , Humans , Recombinant Proteins/isolation & purification
9.
MAbs ; 5(3): 418-31, 2013.
Article in English | MEDLINE | ID: mdl-23567210

ABSTRACT

Optimization of biophysical properties is a critical success factor for the developability of monoclonal antibodies with potential therapeutic applications. The inter-domain disulfide bond between light chain (Lc) and heavy chain (Hc) in human IgG1 lends structural support for antibody scaffold stability, optimal antigen binding, and normal Fc function. Recently, human IgG1λ has been suggested to exhibit significantly greater susceptibility to reduction of the inter Lc-Hc disulfide bond relative to the same disulfide bond in human IgG1κ. To understand the molecular basis for this observed difference in stability, the sequence and structure of human IgG1λ and human IgG1κ were compared. Based on this Lc comparison, three single mutations were made in the λ Lc proximal to the cysteine residue, which forms a disulfide bond with the Hc. We determined that deletion of S214 (dS) improved resistance of the association between Lc and Hc to thermal stress. In addition, deletion of this terminal serine from the Lc of IgG1λ provided further benefit, including an increase in stability at elevated pH, increased yield from transient transfection, and improved in vitro antibody dependent cell-mediated cytotoxicity (ADCC). These observations support the conclusion that the presence of the terminal serine of the λ Lc creates a weaker inter-chain disulfide bond between the Lc and Hc, leading to slightly reduced stability and a potential compromise in IgG1λ function. Our data from a human IgG1λ provide a basis for further investigation of the effects of deleting terminal serine from λLc on the stability and function of other human IgG1λ antibodies.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunoglobulin G/metabolism , Immunoglobulin Light Chains/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibody-Dependent Cell Cytotoxicity/genetics , CHO Cells , Cricetinae , Cricetulus , Cysteine/genetics , HEK293 Cells , Hot Temperature/adverse effects , Humans , Immunoglobulin G/genetics , Immunoglobulin Light Chains/genetics , Mutagenesis, Site-Directed , Mutation/genetics , Protein Binding/genetics , Protein Stability , Serine/genetics
10.
Structure ; 19(8): 1097-107, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21827946

ABSTRACT

The anti-VEGF receptor 2 antibody IMC-1121B is a promising antiangiogenic drug being tested for treatment of breast and gastric cancer. We have determined the structure of the 1121B Fab fragment in complex with domain 3 of VEGFR2, as well as the structure of a different neutralizing anti-VEGFR2 antibody, 6.64, also in complex with VEGFR2 domain 3. The two Fab fragments bind at opposite ends of VEGFR2 domain 3; 1121B directly blocks VEGF binding, whereas 6.64 may prevent receptor dimerization by perturbing the domain 3:domain 4 interface. Mutagenesis reveals that residues essential for VEGF, 1121B, and 6.64 binding are nonoverlapping among the three contact patches.


Subject(s)
Angiogenesis Inhibitors/chemistry , Antibodies, Monoclonal/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry , Amino Acid Motifs , Amino Acid Sequence , Antibodies, Monoclonal, Humanized , Antibody Specificity , Binding Sites, Antibody , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Vascular Endothelial Growth Factor Receptor-2/genetics , Ramucirumab
11.
Hum Antibodies ; 19(4): 89-99, 2010.
Article in English | MEDLINE | ID: mdl-21178280

ABSTRACT

PURPOSE: To evaluate the antibody-dependent cellular cytotoxicity (ADCC) of cetuximab, an anti-epidermal growth factor receptor (EGFR) IgG1 antibody, in vitro. METHODS: Binding to human Fc receptors was measured by ELISA. ADCC against a panel of tumor cell lines was evaluated using peripheral blood mononuclear cells or NK cells as effectors and lactate dehydrogenase release as a marker of cell killing. Cetuximab was compared with two glycan variants of cetuximab and with panitumumab, an anti-EGFR IgG2. RESULTS: Cetuximab bound with high affinity to FcγRI (EC50 = 0.13 nM) and FcγRIIIa (EC50 = 6 nM) and effectively induced ADCC across multiple tumor cell lines. Panitumumab and aglycosylated cetuximab did not bind to FcγRI or FcγRIIIa nor have ADCC activity even at high effector-target cell ratios, even though the EGFR-binding affinity of cetuximab and panitumumab were shown to be comparable (KD = 87 pM and 83 pM, respectively). The extent of cetuximab-elicited ADCC was associated with the level of EGFR expression on tumor cells. CONCLUSIONS: Cetuximab elicits effective ADCC activity against a wide range of tumor cells in vitro. This activity is dependent on antibody glycosylation and IgG1 isotype as well as tumor-cell EGFR expression. These findings suggest that ADCC may contribute to the antitumor activity of cetuximab.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/immunology , ErbB Receptors/metabolism , Immunoglobulin G/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cetuximab , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/immunology , Glycosylation , Humans , Immunoglobulin Isotypes , Killer Cells, Natural/immunology , L-Lactate Dehydrogenase/metabolism , Leukocytes, Mononuclear/immunology , Panitumumab
12.
Clin Cancer Res ; 16(4): 1191-205, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145179

ABSTRACT

PURPOSE: Transforming growth factor beta (TGFbeta) is a pleiotropic cytokine that affects tumor growth, metastasis, stroma, and immune response. We investigated the therapeutic efficacy of anti-TGFbeta receptor II (TGFbeta RII) antibody in controlling metastasis and tumor growth as well as enhancing antitumor immunity in preclinical tumor models. EXPERIMENTAL DESIGN: We generated neutralizing antibodies to TGFbeta RII and assessed the antibody effects on cancer, stroma, and immune cells in vitro. The efficacy and mechanism of action of the antibody as monotherapy and in combination with chemotherapy in suppression of primary tumor growth and metastasis were evaluated in several tumor models. RESULTS: Anti-TGFbeta RII antibody blocked TGFbeta RII binding to TGFbeta 1, 2, and 3, and attenuated the TGFbeta-mediated activation of downstream Smad2 kinase, invasion of cancer cells, motility of endothelial and fibroblast cells, and induction of immunosuppressive cells. Treatment with the antibody significantly suppressed primary tumor growth and metastasis and enhanced natural killer and CTL activity in tumor-bearing mice. Immunohistochemistry analysis showed cancer cell apoptosis and massive necrosis, and increased tumor-infiltrating T effector cells and decreased tumor-infiltrating Gr-1+ myeloid cells in the antibody-treated tumors. Fluorescence-activated cell sorting analysis indicated the significant reduction of peripheral Gr-1+/CD11b+ myeloid cells in treated animals. Concomitant treatment with the cytotoxic agent cyclophosphamide resulted in a significantly increased antitumor efficacy against primary tumor growth and metastasis. CONCLUSIONS: These preclinical data provide a foundation to support using anti-TGFbeta RII antibody as a therapeutic agent for TGFbeta RII-dependent cancer with metastatic capacity.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/pharmacology , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/immunology , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Receptor, Transforming Growth Factor-beta Type II , Smad2 Protein/metabolism , Xenograft Model Antitumor Assays
13.
Neoplasia ; 11(6): 594-604, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19484148

ABSTRACT

Platelet-derived growth factor receptor beta (PDGFRbeta) is upregulated in most of solid tumors. It is expressed by pericytes/smooth muscle cells, fibroblast, macrophage, and certain tumor cells. Several PDGF receptor-related antagonists are being developed as potential antitumor agents and have demonstrated promising antitumor activity in both preclinical and clinical settings. Here, we produced a fully human neutralizing antibody, IMC-2C5, directed against PDGFRbeta from an antibody phage display library. IMC-2C5 binds to both human and mouse PDGFRbeta and blocks PDGF-B from binding to the receptor. IMC-2C5 also blocks ligand-stimulated activation of PDGFRbeta and downstream signaling molecules in tumor cells. In animal studies, IMC-2C5 significantly delayed the growth of OVCAR-8 and NCI-H460 human tumor xenografts in nude mice but failed to show antitumor activities in OVCAR-5 and Caki-1 xenografts. Our results indicate that the antitumor efficacy of IMC-2C5 is primarily due to its effects on tumor stroma, rather than on tumor cells directly. Combination of IMC-2C5 and DC101, an anti-mouse vascular endothelial growth factor receptor 2 antibody, resulted in significantly enhanced antitumor activity in BxPC-3, NCI-H460, and HCT-116 xenografts, compared with DC101 alone, and the trend of additive effects to DC101 treatment in several other tumor models. ELISA analysis of NCI-H460 tumor homogenates showed that IMC-2C5 attenuated protein level of vascular endothelial growth factor and basic fibroblast growth factor elevated by DC101 treatment. Finally, IMC-2C5 showed a trend of additive effects when combined with DC101/chemotherapy in MIA-PaCa-2 and NCI-H460 models. Taken together, these results lend great support to the use of PDGFRbeta antagonists in combination with other antiangiogenic agents in the treatment of a broad range of human cancers.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Receptor, Platelet-Derived Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Cell Line, Tumor , Drug Synergism , Drug Therapy, Combination , Female , Flow Cytometry , HCT116 Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasms/pathology , Peptide Library , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Biochem Biophys Res Commun ; 357(4): 1142-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17462601

ABSTRACT

Platelet-derived growth factor (PDGF) and its receptors (PDGFR) play important roles in tumorigenesis through stimulating tumor growth and promoting angiogenesis via enhancing pericyte recruitment and vessel maturation. Here we produced a neutralizing antibody, 1B3, directed against mouse PDGFRbeta. 1B3 binds to PDGFRbeta with high affinity (9x10(-11)M) and blocks PDGF-BB from binding to the receptor with an IC(50) of approximately 1.2 nM. The antibody also blocks ligand-stimulated activation of PDGFRbeta and downstream signaling molecules, including Akt and MAPK p42/44, in tumor cells. In animal studies, 1B3 significantly enhanced the antitumor and the anti-angiogenic activities of DC101, an antibody directed against mouse vascular endothelial growth factor receptor 2, in a pancreatic (BxPC-3) and a non-small cell lung (NCI-H460) tumor xenograft models. Treatment with the combination of 1B3 and DC101 in BxPC-3 xenograft-bearing mice resulted in tumor regression in 58% of mice compared to that in 18% of mice treated with DC101 alone. Taken together, these results lend great support to use PDGFRbeta antagonists in combinations with other antitumor and/or anti-angiogenic agents in the treatment of a variety of cancers.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Receptor, Platelet-Derived Growth Factor beta/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
15.
J Immunol Methods ; 318(1-2): 65-74, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-17126853

ABSTRACT

Bispecific antibodies (BsAb) have been traditionally utilized to redirect cytotoxic effector cells and agents to kill tumor cells expressing the target antigens. Recently a new concept is emerging to develop BsAb that simultaneously block the functions of two tumor-associated targets, eg., growth factor receptors, for enhanced antitumor efficacies. Broad clinical applications of BsAb have been, and still are, significantly hampered by the difficulty in producing the materials in sufficient quantity and quality by traditional approaches. Here we describe a recombinant approach for the production of an Fc domain-containing, IgG-like tetravalent BsAb, using a single variable domain (sVD) antibody as a versatile building block. In this method, a sVD of a defined specificity is genetically fused to either the N-terminus of the light chain or the C-terminus of the heavy chain of a functional IgG antibody of a different specificity. A model BsAb was constructed using a sVD to mouse platelet derived growth factor receptor alpha and a conventional IgG antibody to mouse platelet derived growth factor receptor beta. The BsAb were expressed in mammalian cells and purified to homogeneity by a one-step Protein A affinity chromatography. Further, the BsAb retained the antigen binding specificity and the receptor neutralizing activity of both of its parent antibodies. Importantly, the BsAb inhibited the activation of both its target receptors in tumor cells stimulated by both platelet derived growth factor AA and BB, whereas the parent monospecific antibody only inhibited the activation of a single receptor stimulated by its cognate ligand. This format of BsAb should be readily applicable to the production of other BsAb recognizing any pairs of antigens.


Subject(s)
Antibodies, Bispecific/immunology , Immunoglobulin Variable Region/genetics , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , Antibody Affinity/immunology , Antibody Specificity/immunology , Becaplermin , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Electrophoresis, Polyacrylamide Gel , Genetic Vectors/genetics , Humans , Immunoglobulin G/immunology , Immunoglobulin Variable Region/immunology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Kinetics , Mice , Models, Molecular , Phosphorylation/drug effects , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/immunology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/immunology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Transfection
16.
Hum Antibodies ; 16(3-4): 127-36, 2007.
Article in English | MEDLINE | ID: mdl-18334748

ABSTRACT

TYRP1 (tyrosinase-related protein 1) is a melanoma antigen expressed in melanosomes and on the surface of melanoma cells. Previous studies have shown that mouse antibodies to TYRP1 localized to melanomas in vivo and inhibited tumor growth and metastasis. Here, we describe the characterization of a novel fully human anti-TYRP1 MAb (20D7) generated by immunizing HuMAb mice (Medarex). 20D7 recognized recombinant and native human TYRP1 by Western blotting and ELISA, and native TYRP1 in melanoma cells as determined by flow cytometry analysis. 20D7 cross-reacted with mouse TYRP1. The binding affinity to human TYRP1 for the human MAb was in the low nM range as determined by surface plasmon resonance kinetics. 20D7 can bind to human and mouse Fc receptor and induce a strong ADCC response against human melanoma cells in vitro. The antitumor activity of 20D7 was tested in human melanoma xenografts and mouse metastatic melanoma models in athymic nude mice. Growth of s.c. human melanoma tumors and metastatic nodules of murine B16 tumor were significantly suppressed by 20D7 compared to human IgG control. These results suggest that human anti-TYRP1 MAb may be a potent therapeutic for the treatment of malignant melanoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Melanoma, Experimental/therapy , Membrane Glycoproteins/immunology , Oxidoreductases/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Complement System Proteins/immunology , Female , Humans , Mice
17.
Clin Cancer Res ; 12(21): 6573-84, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17085673

ABSTRACT

PURPOSE: Vascular endothelial growth factor receptor-1 (VEGFR-1) plays important roles in promotion of tumor growth by mediating cellular functions in tumor vascular endothelium and cancer cells. Blockade of VEGFR-1 activation has been shown to inhibit pathologic angiogenesis and tumor growth, implicating VEGFR-1 as a potential therapeutic target for the treatment of cancer. We have thus developed a VEGFR-1 antagonist human monoclonal antibody designated as IMC-18F1 and evaluated its antitumor activity in preclinical experimental models to show the therapeutic potential of the antibody for cancer treatment in clinic. EXPERIMENTAL DESIGN: Human IgG transgenic mice were used for generation of anti-VEGFR-1 antibodies. Anti-VEGFR-1-specific blocking antibodies were identified using solid-phase binding and blocking assays. Inhibitory antitumor cell activity of IMC-18F1 was assessed in cell-based kinase and growth assays. Pharmacokinetic/pharmacodynamic studies were done to determine the association of antibody blood level with antitumor efficacy of the antibody in vivo. Antitumor efficacy of the anti-VEGFR-1 antibodies as monotherapy and in combination with cytotoxic agents was evaluated in human breast cancer xenograft models. RESULTS: A fully human neutralizing antibody, IMC-18F1, was shown to be a high-affinity (KD=54 pmol) inhibitor of VEGFR-1 ligand binding (VEGF-A, VEGF-B, and placental growth factor). IMC-18F1 inhibited ligand-induced intracellular activation of VEGFR-1 and mitogen-activated protein kinase signaling and prevented ligand-stimulated in vitro growth of breast cancer cells. In vivo, IMC-18F1 suppressed the growth of human breast tumor xenografts in association with reduced mitogen-activated protein kinase and Akt activation, reduced tumor cell proliferation, and increased tumor cell apoptosis. Pharmacokinetic/pharmacodynamic studies established a plasma elimination half-life of 5 days for IMC-18F1 and a steady-state trough plasma therapeutic threshold of 88 microg/mL. Importantly, inhibition of mouse and human VEGFR-1 with MF1 and IMC-18F1, respectively, enhanced the antitumor efficacy of cytotoxic agents commonly used to treat breast cancer. CONCLUSIONS: Based on preclinical validation studies, IMC-18F1 anti-VEGFR-1 has potential to provide clinical benefit to cancer patients.


Subject(s)
Antibodies, Blocking/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Antibodies, Blocking/blood , Antibody Affinity , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blotting, Western , Cell Line, Tumor , Female , Flow Cytometry , Half-Life , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Receptors, Vascular Endothelial Growth Factor/immunology , Xenograft Model Antitumor Assays
18.
Cancer Res ; 66(18): 9162-70, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16982759

ABSTRACT

RON is a member of the c-MET receptor tyrosine kinase family. Like c-MET, RON is expressed by a variety of epithelial-derived tumors and cancer cell lines and it is thought to play a functional role in tumorigenesis. To date, antagonists of RON activity have not been tested in vivo to validate RON as a potential cancer target. In this report, we used an antibody phage display library to generate IMC-41A10, a human immunoglobulin G1 (IgG1) antibody that binds with high affinity (ED50 = 0.15 nmol/L) to RON and effectively blocks interaction with its ligand, macrophage-stimulating protein (MSP; IC50 = 2 nmol/L). We found IMC-41A10 to be a potent inhibitor of receptor and downstream signaling, cell migration, and tumorigenesis. It antagonized MSP-induced phosphorylation of RON, mitogen-activated protein kinase (MAPK), and AKT in several cancer cell lines. In HT-29 colon, NCI-H292 lung, and BXPC-3 pancreatic cancer xenograft tumor models, IMC-41A10 inhibited tumor growth by 50% to 60% as a single agent, and in BXPC-3 xenografts, it led to tumor regressions when combined with Erbitux. Western blot analyses of HT-29 and NCI-H292 xenograft tumors treated with IMC-41A10 revealed a decrease in MAPK phosphorylation compared with control IgG-treated tumors, suggesting that inhibition of MAPK activity may be required for the antitumor activity of IMC-41A10. To our knowledge, this is the first demonstration that a RON antagonist and specifically an inhibitory antibody of RON negatively affects tumorigenesis. Another major contribution of this report is an extensive analysis of RON expression in approximately 100 cancer cell lines and approximately 300 patient tumor samples representing 10 major cancer types. Taken together, our results highlight the potential therapeutic usefulness of RON activity inhibition in human cancers.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/immunology , HT29 Cells , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/metabolism , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NIH 3T3 Cells , Peptide Library , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Xenograft Model Antitumor Assays
19.
Biochem Biophys Res Commun ; 345(1): 438-45, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16682007

ABSTRACT

Compelling evidence suggest that vascular endothelial growth factor (VEGF) and its receptors, especially receptor 2 (VEGFR2, or kinase insert domain-containing receptor, KDR), play a critical role in angiogenesis under both physiological and pathological conditions, including cancer and angiogenic retinopathies such as age-related macular degeneration (AMD). To this end, inhibition of angiogenesis with antagonists to either VEGF or KDR has yielded significant therapeutic efficacy both in preclinical studies in animal models and in clinical trials in patients with cancer and AMD. We previously reported the identification of a high affinity, fully human anti-KDR antibody fragment, 1121B Fab, through a highly stringent affinity maturation process with a Fab originally isolated from a naïve human antibody phage display library. In this study, we demonstrate that 1121B Fab is able to strongly block KDR/VEGF interaction, resulting in potent inhibition of an array of biological activities of VEGF, including activation of the receptor and its signaling pathway, intracellular calcium mobilization, and migration and proliferation of endothelial cells. Taken together, our data lend strong support to the further development of 1121B Fab fragment as an anti-angiogenesis agent in both cancer and angiogenic retinopathies.


Subject(s)
Antibodies, Monoclonal/immunology , Endothelial Cells/immunology , Immunoglobulin Fab Fragments/immunology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Humans , Swine
20.
Mol Cancer Ther ; 4(3): 369-79, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15767546

ABSTRACT

Platelet-derived growth factor receptor alpha (PDGFRalpha) is a type III receptor tyrosine kinase that is expressed on a variety of tumor types. A neutralizing monoclonal antibody to human PDGFRalpha, which did not cross-react with the beta form of the receptor, was generated. The fully human antibody, termed 3G3, has a Kd of 40 pmol/L and blocks both PDGF-AA and PDGF-BB ligands from binding to PDGFRalpha. In addition to blocking ligand-induced cell mitogenesis and receptor autophosphorylation, 3G3 inhibited phosphorylation of the downstream signaling molecules Akt and mitogen-activated protein kinase. This inhibition was seen in both transfected and tumor cell lines expressing PDGFRalpha. The in vivo antitumor activity of 3G3 was tested in human glioblastoma (U118) and leiomyosarcoma (SKLMS-1) xenograft tumor models in athymic nude mice. Antibody 3G3 significantly inhibited the growth of U118 (P=0.0004) and SKLMS-1 (P <0.0001) tumors relative to control. These data suggest that 3G3 may be useful for the treatment of tumors that express PDGFRalpha.


Subject(s)
Antibodies, Monoclonal/chemistry , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Becaplermin , Biological Assay , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Flow Cytometry , Humans , Kinetics , Ligands , MAP Kinase Signaling System , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Phosphorylation , Platelet-Derived Growth Factor/chemistry , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor alpha/immunology , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...