Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Pharm ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941565

ABSTRACT

Lymphocyte activation gene 3 (LAG-3) has attracted much attention as a potentially valuable immune checkpoint. Individual identification of LAG-3 expression at screening and during treatment could improve the successful implementation of anti-LAG-3 therapies. HuL13 is a human IgG1 monoclonal antibody that binds to the LAG-3 receptor in T cells. Here, we used [89Zr]Zr-labeled HuL13 to delineate LAG-3+ T-cell infiltration into tumors via positron emission tomography (PET) imaging. A549/LAG-3 cells, which stably express LAG-3, were generated by infection with lentivirus. The uptake of [89Zr]Zr-DFO-HuL13 in A549/LAG-3 cells was greater than that in the negative control (A549/NC) cells at each time point. The equilibrium dissociation constant (Kd) of [89Zr]Zr-DFO-HuL13 for the LAG-3 receptor was 8.22 nM. PET imaging revealed significant uptake in the tumor areas of A549/LAG-3 tumor-bearing mice from 24 h after injection (SUVmax = 2.43 ± 0.06 at 24 h). As a proof of concept, PET imaging of the [89Zr]Zr-DFO-HuL13 tracer was further investigated in an MC38 tumor-bearing humanized LAG-3 mouse model. PET imaging revealed that the [89Zr]Zr-DFO-HuL13 tracer specifically targets human LAG-3 expressed on tumor-infiltrating lymphocytes (TILs). In addition to the tumors, the spleen was also noticeably visible. Tumor uptake of the [89Zr]Zr-DFO-HuL13 tracer was lower than its uptake in the spleen, but high uptake in the spleen could be reduced by coinjection of unlabeled antibodies. Coinjection of unlabeled antibodies increases tracer activity in the blood pool, thereby improving tumor uptake. Dosimetry evaluation of the healthy mouse models revealed that the highest absorbed radiation dose was in the spleen, followed by the liver and heart wall. In summary, these studies demonstrate the feasibility of using the [89Zr]Zr-DFO-HuL13 tracer for the detection of LAG-3 expression on TILs. Further clinical evaluation of the [89Zr]Zr-DFO-HuL13 tracer may be of significant help in the stratification and management of patients suitable for anti-LAG-3 therapy.

2.
Blood ; 137(6): 844-847, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33181828

ABSTRACT

Agkisacucetin, a snake C-type lectin-like protein isolated from the venom of Deinagkistrodon acutus (formerly Agkistrodon acutus), is a novel antithrombotic drug candidate in phase 2 clinical trials. Agkisacucetin specifically recognizes the platelet surface receptor glycoprotein Ib α chain (GPIbα) to block GPIb and von Willebrand factor (VWF). In this study, we solved the crystal structure of the GPIbα N-terminal domain (residues 1-305) in complex with agkisacucetin to understand their molecular recognition mechanism. The crystal structure showed that agkisacucetin primarily contacts GPIbα at the C-terminal part of the conserved leucine-rich repeat (LRR) domain (LRR-6 to LRR-8) and the previously described "ß-switch" region through the ß chain. In addition, we found that agkisacucetin α chain contacts part of the GPIbα C-terminal peptide after the LRR domain through complementary charge interactions. This C-terminal peptide plays a key role in GPIbα and thrombin recognition. Therefore, our structure revealed that agkisacucetin can sterically block the interaction between the GPIb receptor and VWF and thrombin proteins to inhibit platelet function. Our structural work provides key molecular insights into how an antithrombotic drug candidate recognizes the GPIb receptor to modulate platelet function to inhibit thrombosis.


Subject(s)
Crotalid Venoms/metabolism , Fibrinolytic Agents/metabolism , Lectins, C-Type/metabolism , Platelet Glycoprotein GPIb-IX Complex/chemistry , Crystallography, X-Ray , Humans , Immunoprecipitation , Models, Molecular , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding/drug effects , Protein Conformation , Protein Domains , Protein Interaction Mapping , Structure-Activity Relationship , Surface Plasmon Resonance , Thrombin/metabolism , von Willebrand Factor/metabolism
3.
Cell Mol Biol Lett ; 28(1): 47, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37259060

ABSTRACT

BACKGROUND: Resistance to immune checkpoint inhibitor (ICI) therapy narrows the efficacy of cancer immunotherapy. Although 4-1BB is a promising drug target as a costimulatory molecule of immune cells, no 4-1BB agonist has been given clinical approval because of severe liver toxicity or limited efficacy. Therefore, a safe and efficient immunostimulatory molecule is urgently needed for cancer immunotherapy. METHODS: HK010 was generated by antibody engineering, and the Fab/antigen complex structure was analyzed using crystallography. The affinity and activity of HK010 were detected by multiple in vitro bioassays, including enzyme-linked immunosorbent assay (ELISA), surface plasmon resonance (SPR), flow cytometry, and luciferase-reporter assays. Humanized mice bearing human PD-L1-expressing MC38 (MC38/hPDL1) or CT26 (CT26/hPDL1) tumor transplants were established to assess the in vivo antitumor activity of HK010. The pharmacokinetics (PK) and toxicity of HK010 were evaluated in cynomolgus monkeys. RESULTS: HK010 was generated as an Fc-muted immunoglobulin (Ig)G4 PD-L1x4-1BB bispecific antibody (BsAb) with a distinguished Fab/antigen complex structure, and maintained a high affinity for human PD-L1 (KD: 2.27 nM) and low affinity for human 4-1BB (KD: 493 nM) to achieve potent PD-1/PD-L1 blockade and appropriate 4-1BB agonism. HK010 exhibited synergistic antitumor activity by blocking the PD-1/PD-L1 signaling pathway and stimulating the 4-1BB signaling pathway simultaneously, and being strictly dependent on the PD-L1 receptor in vitro and in vivo. In particular, when the dose was decreased to 0.3 mg/kg, HK010 still showed a strong antitumor effect in a humanized mouse model bearing MC38/hPDL1 tumors. Strikingly, HK010 treatment enhanced antitumor immunity and induced durable antigen-specific immune memory to prevent rechallenged tumor growth by recruiting CD8+ T cells and other lymphocytes into tumor tissue and activating tumor-infiltrating lymphocytes. Moreover, HK010 not only did not induce nonspecific production of proinflammatory cytokines but was also observed to be well tolerated in cynomolgus monkeys in 5 week repeated-dose (5, 15, or 50 mg/kg) and single-dose (75 or 150 mg/kg) toxicity studies. CONCLUSION: We generated an Fc-muted anti-PD-L1x4-1BB BsAb, HK010, with a distinguished structural interaction with PD-L1 and 4-1BB that exhibits a synergistic antitumor effect by blocking the PD-1/PD-L1 signaling pathway and stimulating the 4-1BB signaling pathway simultaneously. It is strictly dependent on the PD-L1 receptor with no systemic toxicity, which may offer a new option for cancer immunotherapy.


Subject(s)
Antibodies, Bispecific , Colorectal Neoplasms , Programmed Cell Death 1 Receptor , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Immunotherapy , Macaca fascicularis , Antibodies, Bispecific/pharmacology
4.
J Transl Med ; 20(1): 415, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36076251

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignancies and the patient survival rate remains unacceptably low. The anti-programmed cell death-1 (PD-1)/programmed cell death ligand 1 (PD-L1) antibody-based immune checkpoint inhibitors have been added to CRC treatment regimens, however, only a fraction of patients benefits. As an important co-stimulatory molecule, 4-1BB/CD137 is mainly expressed on the surface of immune cells including T and natural killer (NK) cells. Several agonistic molecules targeting 4-1BB have been clinically unsuccessful due to systemic toxicity or weak antitumor effects. We generated a humanized anti-4-1BB IgG4 antibody, HuB6, directed against a unique epitope and hypothesized that it would promote antitumor immunity with high safety. METHODS: The antigen binding specificity, affinity and activity of HuB6 were determined by enzyme-linked immunosorbent assay (ELISA), surface plasmon resonance (SPR), biolayer interferometry (BLI) and flow cytometry. The antitumor effects were evaluated in humanized mice bearing syngeneic tumors, and possible toxicity was evaluated in humanized mice and cynomolgus monkeys. RESULTS: HuB6 showed high specificity and affinity for a binding epitope distinct from those of other known 4-1BB agonists, including utomilumab and urelumab, and induced CD8 + T, CD4 + T and NK cell stimulation dependent on Fcγ receptor (FcγR) crosslinking. HuB6 inhibited CRC tumor growth in a dose-dependent manner, and the antitumor effect was similar with urelumab and utomilumab in humanized mouse models of syngeneic CRC. Furthermore, HuB6 combined with an anti-PD-L1 antibody significantly inhibited CRC growth in vivo. Additionally, HuB6 induced antitumor immune memory in tumor model mice rechallenged with 4 × 106 tumor cells. Toxicology data for humanized 4-1BB mice and cynomolgus monkeys showed that HuB6 could be tolerated up to a 180 mg/kg dose without systemic toxicity. CONCLUSIONS: This study demonstrated that HuB6 should be a suitable candidate for further clinical development and a potential agent for CRC immunotherapy.


Subject(s)
Colorectal Neoplasms , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Colorectal Neoplasms/drug therapy , Epitopes , Immunotherapy , Macaca fascicularis , Mice , Receptors, IgG
5.
Biochem J ; 477(19): 3833-3838, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32969463

ABSTRACT

Post-translational modifications play important roles in mediating protein functions in a wide variety of cellular events in vivo. HEMK2-TRMT112 heterodimer has been reported to be responsible for both histone lysine methylation and eukaryotic release factor 1 (eRF1) glutamine methylation. However, how HEMK2-TRMT112 complex recognizes and catalyzes eRF1 glutamine methylation is largely unknown. Here, we present two structures of HEMK2-TRMT112, with one bound to SAM and the other bound with SAH and methylglutamine (Qme). Structural analyses of the post-catalytic complex, complemented by mass spectrometry experiments, indicate that the HEMK2 utilizes a specific pocket to accommodate the substrate glutamine and catalyzes the subsequent methylation. Therefore, our work not only throws light on the protein glutamine methylation mechanism, but also reveals the dual activity of HEMK2 by catalyzing the methylation of both Lys and Gln residues.


Subject(s)
Glutamine/chemistry , Methyltransferases/chemistry , Site-Specific DNA-Methyltransferase (Adenine-Specific)/chemistry , Glutamine/metabolism , Humans , Methylation , Methyltransferases/metabolism , Protein Structure, Quaternary , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism
6.
Am J Cancer Res ; 13(7): 3246-3256, 2023.
Article in English | MEDLINE | ID: mdl-37559991

ABSTRACT

Resistance to HER2-targeted therapy narrows the efficacy of cancer immunotherapy. Although 4-1BB/CD137 is a promising drug target as a costimulatory molecule of immune cells, no therapeutic drug has been approved in the clinic because of systemic toxicity or limited efficacy. Previously, we developed a humanized anti-HER2 monoclonal antibody (mAb) HuA21 and anti-4-1BB mAb HuB6 with distinct antigen epitopes for cancer therapy. Here, we generated an Fc-muted IgG4 HER2/4-1BB bispecific antibody (BsAb) HK006 by the fusion of HuB6 scFv and HuA21 Fab. HK006 exhibited synergistic antitumor activity by blocking HER2 signal transduction and stimulating the 4-1BB signaling pathway simultaneously and strictly dependent on HER2 expression in vitro and in vivo. Strikingly, HK006 treatment enhanced antitumor immunity by increasing and activating tumor-infiltrating T cells. Moreover, HK006 did not induce nonspecific production of proinflammatory cytokines and had no obvious toxicity in mice. Overall, these data demonstrated that HK006 should be a promising candidate for HER2-positive cancer immunotherapy.

7.
J Biol Chem ; 286(36): 31676-83, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21680730

ABSTRACT

p185(her2/neu) belongs to the ErbB receptor tyrosine kinase family, which has been associated with human breast, ovarian, and lung cancers. Targeted therapies employing ectodomain-specific p185(her2/neu) monoclonal antibodies (mAbs) have demonstrated clinical efficacy for breast cancer. Our previous studies have shown that p185(her2/neu) mAbs are able to disable the kinase activity of homomeric and heteromeric kinase complexes and induce the conversion of the malignant to normal phenotype. We previously developed a chimeric antibody chA21 that specifically inhibits the growth of p185(her2/neu)-overexpressing cancer cells in vitro and in vivo. Herein, we report the crystal structure of the single-chain Fv of chA21 in complex with an N-terminal fragment of p185(her2/neu), which reveals that chA21 binds a region opposite to the dimerization interface, indicating that chA21 does not directly disrupt the dimerization. In contrast, the bivalent chA21 leads to internalization and down-regulation of p185(her2/neu). We propose a structure-based model in which chA21 cross-links two p185(her2/neu) molecules on separate homo- or heterodimers to form a large oligomer in the cell membrane. This model reveals a mechanism for mAbs to drive the receptors into the internalization/degradation path from the inactive hypophosphorylated tetramers formed dynamically by active dimers during a "physiologic process."


Subject(s)
Antibodies, Monoclonal/chemistry , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Crystallography, X-Ray , Down-Regulation/immunology , Endocytosis , Humans , Receptor, ErbB-2/genetics
8.
Cancer Immunol Immunother ; 60(3): 339-48, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21086124

ABSTRACT

It was well studied that ErbB2 (HER2/p185(her2/neu)) overexpression in human malignant cancers correlates with poor prognosis and chemo-resistance. Although Trastuzumab (Herceptin) has been widely used in patients with ErbB2-overexpressing metastatic breast cancer, many patients either do not respond to Trastuzumab therapy or progress within 1 year of initiating Trastuzumab treatment. Previously, we reported a novel tumor-inhibitory antibody chA21, which recognized ErbB2 extracellular domain with an epitope distinct from other tumor-inhibitory anti-ErbB2 antibodies. Here, we report that chA21 combined with Paclitaxel or Trastuzumab significantly enhances the tumor-inhibition effects on ErbB2-overexpressing breast and ovarian cancer in xenograft mice. Moreover, the study reveals that the effects by chA21 to cause an enhanced inhibition on cancer cell proliferation and angiogenesis was highly associated with the intrinsic ability of chA21 to down-regulate ErbB2 receptor, inhibit downstream MAPK and PI3K-AKT signal transduction and activate natural killer cells. Our findings show that chA21 may represent a unique anti-ErbB2 antibody with potentials as therapeutic candidate alone or combination with other anti-ErbB2 reagents in cancer therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Paclitaxel/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Trastuzumab , Xenograft Model Antitumor Assays
9.
Acta Crystallogr D Struct Biol ; 75(Pt 6): 554-563, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-31205018

ABSTRACT

HER2, a member of the epidermal growth factor receptor (EGFR) family, has been associated with human breast, ovarian and gastric cancers. Anti-HER2 monoclonal antibodies (mAbs) have demonstrated clinical efficacy for HER2-overexpressing breast cancer. A chimeric antibody chA21 that specifically inhibits the growth of HER2-overexpressing cancer cells both in vitro and in vivo has previously been developed. To reduce a potential human anti-mouse immune response, the humanized antibody HuA21 was developed and was further subjected to affinity maturation by phage display on the basis of chA21. Here, the crystal structure of HuA21-scFv in complex with the extracellular domain of HER2 is reported, which demonstrates that HuA21 binds almost the same epitope as chA21 and also provides insight into how substitutions in HuA21 improve the binding affinity compared with chA21, which could facilitate structure-based optimization in the future. Furthermore, the effects of HuA21 variants with constant domains of different lengths were explored and it was noticed that the deletion of constant domain 1 could improve the inhibition efficacy in a cell-proliferation assay, possibly functioning via increased internalization, which might guide the design of other monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antigen-Antibody Complex/chemistry , Antineoplastic Agents, Immunological/chemistry , Neoplasms/therapy , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Binding Sites, Antibody , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Crystallization , Crystallography, X-Ray/methods , Female , Humans , Protein Domains , Receptor, ErbB-2/immunology
10.
Biochim Biophys Acta ; 1769(9-10): 593-602, 2007.
Article in English | MEDLINE | ID: mdl-17822787

ABSTRACT

HEC1 (highly expressed in cancer), which localizes to kinetochore in cell mitosis, plays an essential role in chromosome segregation for M phase progression. To clarify the mechanism of its transcriptional regulation, we searched out and isolated its 5'-flanking region. Mapping of this region identified that it is a TATA-less promoter and contains several putative binding sites for different transcription factors. The results from HeLa cells transfected with pGL3 luciferase reporter vectors containing progressive deletion of the HEC1 5'-flanking region demonstrated that two elements containing binding sites for cAMP responsive element binding (CREB) protein and activating transcription factor 4 (ATF4 or CREB2) are critical for transcriptional activity. Mutation of the two elements, not downstream E2F box, resulted in a significant reduction of the promoter activity. Gel shift and supershift assays also demonstrated specific binding of transcription factors to their putative binding sites. Furthermore, overexpression of either CREB or ATF4 enhanced the activation of the HEC1 promoter and overexpression of both of them had an additive effect on the activation of the HEC1 transcription. Conversely, overexpression of dominant negative mutants of either CREB or ATF4 resulted in downregulation of HEC1 mRNA significantly. Our study provided a new insight into a potential mechanism of how transcription factors of CREB family are involved in the regulation of kinetochore protein HEC1 in cancer-related cells.


Subject(s)
Activating Transcription Factor 4/metabolism , Cell Cycle Proteins/biosynthesis , Cell Division/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/physiology , Nuclear Proteins/biosynthesis , Transcription, Genetic/physiology , Activating Transcription Factor 4/genetics , Cell Cycle Proteins/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cytoskeletal Proteins , Down-Regulation/genetics , HeLa Cells , Humans , Kinetochores/metabolism , Mutation , Nuclear Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Response Elements/physiology
11.
Proteins ; 70(3): 938-49, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17847085

ABSTRACT

Anti-ErbB2 antibodies targeting distinct epitopes can have different biological functions on cancer cells. A21 prepared by surface epitope masking (SEM) method is a tumor-inhibitory anti-ErbB2 monoclonal antibody. Previously we engineered a single chain chimeric antibody chA21 with potential for therapy of ErbB2-overexpressing tumors. Here, we mapped the A21 epitope on ErbB2 extracellular domain (ECD) by screening a combinatorial phage display peptide library, serial subdomain deletion, and mutagenesis scanning. X-ray crystal structure of the A21 scFv fragment at 2.1 A resolution was also determined. A molecular model of Ag-Ab complex was then constructed based on the crystal structures of the A21 scFv and ErbB2 ECD. Some of biological functions of the A21 mAb and its derivative antibodies including their tumor cell growth inhibition and effects on the expression, internalization, and phosphorylation of ErbB2 receptor were also investigated. The results showed that A21 recognized a conformational epitope comprising a large region mostly from ErbB2 extracellular subdomain I with several surface-exposed residues important for the binding affinity. These data provide unique functional properties of A21 that are quite different from two broadly used anti-ErbB2 mAbs, Herceptin and 2C4. It suggested that the A21 epitope may be another valuable target for designing new anti-ErbB2 therapeutics.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Epitopes/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antineoplastic Agents/pharmacology , Binding Sites, Antibody , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Epitope Mapping , Epitopes/immunology , Epitopes/metabolism , Female , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Models, Molecular , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured
12.
J Exp Clin Cancer Res ; 29: 23, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20214830

ABSTRACT

BACKGROUND AND AIMS: Anti-HER-2 antibodies targeting distinct epitopes have different biological functions on cancer cells. In a previous study, we demonstrated that anti-HER-2 engineering antibody ChA21 was able to bind to subdomain I of HER-2 extracellular domain. In this study, The effects of ChA21 on growth and apoptosis against ovarian carcinoma cell SK-OV-3 over-expressing HER-2 in vitro and in vivo were investigated. METHODS: Cell growth inhibition was evaluated by MTT assay. Apoptosis was detected by TUNEL stain, transmission electron microscopy and flow cytometry on cultured cells and tissue sections from nude mice xenografts. The apoptosis-related proteins Bax and Bcl-2 were assessed by immunohistochemistry. RESULTS: We found that treatment of ChA21 caused a dose-dependent decrease of cell proliferation in vitro and a significant inhibition of tumor growth in vivo. ChA21 therapy led to a significant increase in the induction of apoptosis, and up-regulated the expression of Bax, while the expression of Bcl-2 was down-regulated. CONCLUSION: These data suggest that ChA21 inhibits the growth and induces apoptosis of SK-OV-3 via regulating the balance between Bax and Bcl-2.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/metabolism
13.
Cancer Lett ; 280(1): 50-60, 2009 Jul 18.
Article in English | MEDLINE | ID: mdl-19269083

ABSTRACT

HRG-beta1 stimulation of breast cancer cell line SKBR3 resulted in not only increased cell migration and invasion, upregulation of some mesenchymal markers, and downregulation of epithelial marker, but also upregulation of transcription factor Snail and its nuclear translocation. Similar results were acquired for cells transfected with Snail cDNA. Furthermore, downregulation of Snail by siRNA attenuated HRG-beta1 induced EMT-like phenotype. Inhibition of Akt kinase activation by a PI3K inhibitor LY294002, or exogenous expression of a kinase-dead mutant of Akt abrogated the increase of Snail expression induced by HRG-beta1. Conversely, expression of a constitutively active Akt resulted in increase of Snail expression. These results indicated that Snail upregulation by HRG-beta1 is mediated via the PI3K/Akt signaling pathway and that Snail plays a key role in HRG-beta1 induced breast cancer cell metastasis through induction of EMT.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Mesoderm/metabolism , Neuregulin-1/physiology , Transcription Factors/biosynthesis , Cell Line, Tumor , Cell Movement , Chemotaxis , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Epithelium/pathology , Humans , Mesoderm/pathology , Morpholines/pharmacology , Neoplasm Metastasis , Neuregulin-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Snail Family Transcription Factors
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 23(3): 260-3, 2007 Mar.
Article in Zh | MEDLINE | ID: mdl-17343796

ABSTRACT

AIM: To explore the apoptotic effect of the combined treatment of anti-p185(c-erbB-2/neu) engineered antibody with paclitaxel on p185-overexpressing human malignant breast cancer cell lines BT474 and to study its emerging mechanism. METHODS: The prohibitory effect of engineeded antibody plus paclitaxel on BT474 cells was assessed by MTS assay. The number of apoptotic cells was detected by Annexin V-FITC/PI. DNA content and cell cycle distribution were determined by FCM; DNA-binding activity of NF-kappaB was demonstrated by EMSA. RESULTS: Anti-p185(c-erbB-2/neu) engineered antibody plus paclitaxel resulted in synergistic effect on proliferative inhibiton of BT474 cells, which was mediated via apoptotic induction and caused cell cycle to arrest at G1 phase remarkably. Furthermore, the combined treatment of the engineered antibody with paclitaxel effectively suppressed the activation of NF-kappaB in BT474 cells. CONCLUSION: The combined treatment of anti-p185(c-erbB-2/neu) engineered antibody with paclitaxel rendered p185-overexpressing human malignant breast cancer cells BT474 more susceptible to paclitaxel-induced apoptosis by the effective suppression of NF-kappaB activation.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Paclitaxel/pharmacology , Antibodies/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Electrophoretic Mobility Shift Assay , Flow Cytometry , G1 Phase/drug effects , Humans , NF-kappa B/metabolism , Receptor, ErbB-2/immunology
15.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 23(8): 691-5, 2007 Aug.
Article in Zh | MEDLINE | ID: mdl-17618556

ABSTRACT

AIM: To explore the possibility of RNA interference (RNAi)-based gene therapy against HER2-overexpressing tumors using adenovirus-mediated vector. METHODS: A plasmid named pHER2-GFP containing HER2 and green fluorescent protein (GFP) fusion was constructed and cotransfected into CHO-K1 cells respectively with nine small interference RNA (siRNA)-expressing plasmids targeting different regions of HER2. The siRNA-expressing plasmids with best interference effect were screened out and then used to identify the gene silence effect in HER2-overexpressing SKBR3 breast cancer cells. Subsequently, the siRNA-expressing cassettes were subcloned into adenoviral vectors. Downregulation of HER2 by adenovirus-mediated RNAi and its effect on SKBR3 cell proliferation were identified again. RESULTS: Two siRNA-expressing plasmids with best interference effect were screened out and HER2 was also efficiently downregulated in SKBR3 cells infected with the adenovirus containing these siRNA-expressing cassettes. Downregulation of HER2 resulted in the increase of cells in G1 phase and the induction of apoptosis. Furthermore, infection of adenovirus inhibited SKBR3 cell growth, which was confirmed by MTT and cell long-term proliferation assays. CONCLUSION: The adenovirus-mediated RNAi could downregulate the HER2 expression efficiently and exert an inhibitory effect on growth of HER2-overexpressing breast cancer cell.


Subject(s)
Down-Regulation , RNA Interference/physiology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Animals , Blotting, Western , CHO Cells , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cricetinae , Cricetulus , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Polymerase Chain Reaction
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 23(11): 1031-3, 2007 Nov.
Article in Zh | MEDLINE | ID: mdl-17988585

ABSTRACT

AIM: To prepare and identify the antibodies against snake venom C-type lectin like protein Agkisacutacin. METHODS: A BALB/c mouse was immunized with Agkisacutacin and the cells were fused by standard hybridoma technique to prepare mAbs. The stability of the hybridomas secreting mAbs was detected by indirect ELISA. The nuclear type of the hybridomas was analyzed by fluorescent staining and the specificity of mAbs was detected by Western blot. RESULTS: Three cell strains of hybridomas that could steadily secrete anti-Agkisacutacin mAb were obtained. CONCLUSION: The successful preparation of anti-Agkisacutacin mAbs provides an important tool for studying the in vivo metabolism of new anti-thrombus drugs by detecting Agkisacutacin and investigating the mechanism of anti-thrombus of the drugs.


Subject(s)
Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Crotalid Venoms/immunology , Snake Venoms/chemistry , Animals , Antibodies, Monoclonal/metabolism , Antibody Specificity , Crotalid Venoms/biosynthesis , Crotalid Venoms/isolation & purification , Enzyme-Linked Immunosorbent Assay , Female , Hybridomas/metabolism , Mice , Mice, Inbred BALB C , Snake Venoms/immunology
17.
Protein Expr Purif ; 47(1): 249-57, 2006 May.
Article in English | MEDLINE | ID: mdl-16403645

ABSTRACT

Anti-ErbB2 antibodies are used as convenient tools in exploration of ErbB2 functional mechanisms and in treatment of ErbB2-overexpressing tumors. When we employed the yeast Pichia pastoris to express an anti-ErbB2 single-chain antibody (scFv) derived from the tumor-inhibitory monoclonal antibody A21, the yield did not exceed 1-2 mg/L in shake flask cultures. As we considered that the poor codon usage bias may be one limiting factor leading to the inefficient translation and scFv production, we designed and synthesized the full-length scFv gene by choosing the P. pastoris preferred codons while keeping the G+C content at relatively low level. Codon optimization increased the scFv expression level 3- to 5-fold and up to 6-10 mg/L. Northern blotting further confirmed that the increase of scFv expression was mainly due to the enhancement of translation efficiency. Investigation of culture conditions revealed that the maximal cell growth and scFv expression were achieved at pH 6.5-7.0 with 2% casamino acids after 72 h methanol induction. Secreted scFv was easily purified (>95% homogeneous product) from culture supernatants in one step by using Ni2+ chelating affinity chromatography. The yield was approximately 10-15 mg/L. Functional studies showed that the A21 scFv could be internalized with high efficiency after binding to the ErbB2-overexpressing cells, suggesting this regent may prove especially useful for ErbB2-targeted immunotherapy.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Cloning, Molecular , Codon/biosynthesis , Pichia , Receptor, ErbB-2/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Base Sequence , Cell Line, Tumor , Codon/chemistry , Humans , Immunoglobulin Fragments/biosynthesis , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/biosynthesis , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Mice , Molecular Sequence Data , Pichia/genetics
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 22(1): 51-3, 57, 2006 Jan.
Article in Zh | MEDLINE | ID: mdl-16388745

ABSTRACT

AIM: To explore the effects of anti-HER-2 chimeric antibody chA21 on proliferation and apoptosis of ovarian cancer cell lines SKOV3. METHODS: MTT colorometric assay, HE staining, transmission electron microscopy, flow cytometry and TUNEL staining were used to study the proliferative inhibition and apoptotic induction of SKOV3 cells by chA21 in vitro. RESULTS: Proliferative inhibition rate and apoptotic rate of SKOV3 cells were increased with dose and action time by chA21 (0.2 mg/L-5.4 mg/L). CONCLUSION: chA21 can remarkably inhibit proliferation of SKOV3 cells in vitro and induction of apoptosis may be a principal way.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Ovarian Neoplasms/drug therapy , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/pharmacology , Cell Line, Tumor , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Microscopy, Electron, Transmission , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure
19.
Protein Expr Purif ; 41(1): 68-76, 2005 May.
Article in English | MEDLINE | ID: mdl-15802223

ABSTRACT

Monoclonal antibody A21 reacts specifically with the extracellular domain of p185c-erbB-2 oncoprotein, a member of the epidermal growth factor receptor family. In a previous study, we constructed a single-chain chimeric antibody, assembled using an A21 single-chain Fv antibody and a human IgG1 Fc fragment. In this study, we expressed this chimeric antibody using a CHO-GS system, and developed a simple and efficient method for its purification. After only one step using affinity purification, the recovery rate and purity of the antibody attained was 60 and 91%, respectively. After a second step, using reverse phase HPLC purification, the purity was above 99%. The high purity of the recombinant antibody allowed us to identify a number of its intrinsic molecular properties, including antigen binding activity, measurement of affinity constant, N-terminal sequencing, and mass spectrometer analysis. These results further augment the potential of this recombinant antibody to be a drug candidate for cancer therapy.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Receptor, ErbB-2/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Binding, Competitive , CHO Cells , Chromatography, Affinity , Chromatography, High Pressure Liquid , Cricetinae , Gene Expression , Humans , In Vitro Techniques , Mice , Molecular Sequence Data , Neoplasms/therapy , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
20.
Sheng Wu Gong Cheng Xue Bao ; 21(4): 590-6, 2005 Jul.
Article in Zh | MEDLINE | ID: mdl-16176098

ABSTRACT

Transmembrane protein p185 (the product of Her2/c-erbB-2 gene) is a member of the epidermal growth factor receptor (EGFR) family. Its overexpression was found in about 30% of breast cancer. It is essential to obtain soluble extracellular domain (ECD) of p185, especially disulfide bond rich domains, for identifying the epitopes of anti-p185 antibodies and researching the interrelationship between the antigen and antibody. The disulfide bond rich domain I-II and domain IV of p185 ECD were amplified from plasmid pBabe/erbB-2 by PCR respectively. These two fragments were inserted into pGEX/4T-1 vector, transfected into E. coli Origami B (DE3) pLysS and expressed inductively by low concentration of IPTG and low temperature overnight. After the pressure lysis of cells, the supernatants were analyzed by SDS-PAGE and the result demonstrated that this GST-fusion protein was expressed solubly in the amount of 10-15 mg/L. By the ELISA, Western blot and other immunological assays, the fusion proteins and their GST cut-off derivates both showed binding activities with several anti-p185 antibodies respectively. These results indicated that it was a feasible and effectual method to express disulfide bond rich domain I-II and domain IV of p185 ECD and this method may also be used to express other disulfide bond rich proteins.


Subject(s)
Disulfides/immunology , Escherichia coli/metabolism , Receptor, ErbB-2/biosynthesis , Antibodies, Monoclonal/immunology , Escherichia coli/genetics , Genetic Vectors/genetics , Humans , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Solubility , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL