Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Int J Mol Sci ; 19(2)2018 Feb 18.
Article in English | MEDLINE | ID: mdl-29463039

ABSTRACT

Epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) resistance is one of the most important problems in lung cancer therapy. Lung adenocarcinoma with EGFR-TKI resistance was reported to have higher abilities of invasion and migration than cancers sensitive to EGFR-TKI, but the function of matrix metalloproteinases (MMPs) has not been explored in EGFR-TKI-resistant lung adenocarcinoma. This study aims to clarify the significance of MMP-1 in EGFR-TKI-resistant lung adenocarcinoma. From the results of in vitro studies of migration and invasion assays using EGFR-TKI-sensitive and -resistant cell lines and phosphorylation antibody arrays using EGF and rapamycin, we first demonstrate that overexpression of MMP-1, which might follow activation of a mammalian target of rapamycin (mTOR) pathway, plays an important role in the migration and invasion abilities of EGFR-TKI-resistant lung adenocarcinoma. Additionally, immunohistochemical studies using 89 cases of lung adenocarcinoma demonstrate that high expression of MMP-1 is significantly correlated with poor prognosis and factors such as smoking history and the subtype of invasive mucinous adenocarcinoma. These are consistent with the results of this in vitro study. To conclude, this study provides insights into the development of a possible alternative therapy manipulating MMP-1 and the mTOR signaling pathway in EGFR-TKI-resistant lung adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Matrix Metalloproteinase 1/metabolism , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aged , Cell Line, Tumor , Cell Movement/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 1/genetics , Multivariate Analysis , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
2.
J Pathol ; 234(2): 277-88, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24687913

ABSTRACT

The development of therapeutic resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs, ie erlotinib or gefitinib) has been the major clinical problem when treating lung adenocarcinoma patients with these agents. However, its mechanisms have not necessarily been well studied to this date. Autophagy has been recently considered to play pivotal roles in escaping from the effects of anti-neoplastic agents. Therefore, in this study, we examined its roles in the development of resistance to EGFR-TKIs in lung adenocarcinoma. We first established erlotinib-resistant cell lines (PC9/ER) from parental PC9 cells by exposing the cells to erlotinib. In PC9/ER, autophagy-related LC3A expression came to be up-regulated and constitutive activation of LC3A-mediated autophagy became more pronounced through the process of acquiring therapeutic resistance. In addition, inhibition of LC3A or autophagy restores sensitivity to EGFR-TKIs in PC9/ER. LC3A was also activated at the transcriptional level in de novo resistant cells via demethylation of the MAP1LC3A gene. We then evaluated the status of LC3A in 169 lung adenocarcinoma patients using immunohistochemistry. LC3A immunoreactivity was only detected in carcinoma cells (89/169 patients), not in non-tumoural cells. In addition, LC3A immunoreactivity was significantly correlated with progression-free survival (p = 0.0039) and overall survival (p = 0.0040) of 35 patients treated with EGFR-TKIs. The results of our present study demonstrated that LC3A-mediated autophagy in carcinoma cells was involved in the development of resistance to EGFR-TKIs, and that LC3A could serve as a promising therapeutic target for overcoming resistance to EGFR-TKIs and a novel predictor of response to EGFR-TKIs in lung adenocarcinoma patients.


Subject(s)
Adenocarcinoma/genetics , Antineoplastic Agents/pharmacology , Autophagy/genetics , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Lung Neoplasms/genetics , Microtubule-Associated Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Erlotinib Hydrochloride , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology
3.
Stem Cells ; 30(12): 2631-44, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23081779

ABSTRACT

The cancer stem cell (CSC) concept has been proposed as an attractive theory to explain cancer development, and CSCs themselves have been considered as targets for the development of diagnostics and therapeutics. However, many unanswered questions concerning the existence of slow cycling/quiescent, drug-resistant CSCs remain. Here we report the establishment of colon cancer CSC lines, interconversion of the CSCs between a proliferating and a drug-resistant state, and reconstitution of tumor hierarchy from the CSCs. Stable cell lines having CSC properties were established from human colon cancer after serial passages in NOD/Shi-scid, IL-2RƎĀ³(null) (NOG) mice and subsequent adherent cell culture of these tumors. By generating specific antibodies against LGR5, we demonstrated that these cells expressed LGR5 and underwent self-renewal using symmetrical divisions. Upon exposure to irinotecan, the LGR5(+) cells transitioned into an LGR5(-) drug-resistant state. The LGR5(-) cells converted to an LGR5(+) state in the absence of the drug. DNA microarray analysis and immunohistochemistry demonstrated that HLA-DMA was specifically expressed in drug-resistant LGR5(-) cells, and epiregulin was expressed in both LGR5(+) and drug-resistant LGR5(-) cells. Both cells sustained tumor initiating activity in NOG mice, giving rise to a tumor tissue hierarchy. In addition, anti-epiregulin antibody was found to be efficacious in a metastatic model. Both LGR5(+) and LGR5(-) cells were detected in the tumor tissues of colon cancer patients. The results provide new biological insights into drug resistance of CSCs and new therapeutic options for cancer treatment.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/biosynthesis , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/immunology , Antibody Specificity , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Colonic Neoplasms/therapy , Drug Resistance, Neoplasm , Epidermal Growth Factor/immunology , Epiregulin , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, G-Protein-Coupled/immunology , Transplantation, Heterologous
4.
Sci Transl Med ; 15(684): eabq5858, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36812343

ABSTRACT

Current pharmacological treatments for endometriosis are limited to hormonal agents that can relieve pain but cannot cure the disease. Therefore, the development of a disease-modifying drug for endometriosis is an unmet medical need. By studying human endometriotic samples, we found that the progression of endometriosis was associated with the development of inflammation and fibrosis. In addition, IL-8 expression was highly up-regulated in endometriotic tissues and closely correlated with disease progression. We created a long-acting recycling antibody against IL-8 (AMY109) and evaluated its clinical potency. Because rodents do not produce IL-8 and do not experience menstruation, we analyzed the lesions in cynomolgus monkeys that spontaneously developed endometriosis and in a surgically induced endometriosis monkey model. Both spontaneously developed and surgically induced endometriotic lesions demonstrated pathophysiology that was highly similar to that of human endometriosis. Once-a-month subcutaneous injection of AMY109 to monkeys with surgically induced endometriosis reduced the volume of nodular lesions, lowered the Revised American Society for Reproductive Medicine score as modified for monkeys, and ameliorated fibrosis and adhesions. In addition, experiments using cells derived from human endometriosis revealed that AMY109 inhibited the recruitment of neutrophils to endometriotic lesions and the production of monocyte chemoattractant protein-1 from neutrophils. Thus, AMY109 may represent a disease-modifying therapy for patients with endometriosis.


Subject(s)
Endometriosis , Female , Humans , Endometriosis/drug therapy , Inflammation , Fibrosis
5.
Int J Cancer ; 128(9): 2215-23, 2011 May 01.
Article in English | MEDLINE | ID: mdl-20602340

ABSTRACT

Parathyroid hormone-related protein (PTHrP) is a causative factor of humoral hypercalcemia in malignancy. However, it is difficult to explain the mechanism of anorexia/cachexia with PTHrP secretion in detail. Previously, we demonstrated that the expressions of orexigenic peptides increased and anorexigenic peptides decreased under cachectic conditions in rats carrying tumors secreting PTHrP. In this study, we investigated whether such changes in the expression of hypothalamic feeding-regulating peptides can be solely attributed to PTHrP or are a general response under cachectic conditions. Cachectic syndromes were induced in rats by: (i) inoculation of human lung cancer LC-6 cells that secreted PTHrP, (ii) inoculation of human melanoma SEKI cells that secrete not PTHrP but LIF1, (iii) injection of heat-killed Mycobacterium leading to arthritis (AA) and (iv) oral administration of a high dose of 1α,25(OH)(2)D(3) that resulted in hypercalcemia. The LC-6-bearing rats and AA rats were treated with or without anti-PTHrP antibody and indomethacin, respectively, and the expression of the hypothalamic feeding-regulating peptide mRNAs were examined by in situ hybridization histochemistry. The orexigenic peptide mRNAs, such as neuropeptide Y and agouti-related protein, were significantly increased, and that of anorexigenic peptide mRNAs, such as proopiomelanocortin, cocaine- and amphetamine-regulated transcript and corticotropin-releasing hormone were significantly decreased when they developed cachectic syndromes and AA. A high dose of 1α,25(OH)(2)D(3) caused hypercalcemia and body weight loss but did not affect the expression of hypothalamic feeding-regulating peptide mRNAs. The expressions of the hypothalamic feeding-regulating peptides change commonly in different chronic cachectic models without relating to serum calcium levels.


Subject(s)
Arthritis, Experimental/metabolism , Cachexia/metabolism , Hypothalamus/metabolism , Leptin/blood , Leukemia Inhibitory Factor/metabolism , Neoplasms, Experimental/metabolism , Parathyroid Hormone-Related Protein/metabolism , Agouti-Related Protein/biosynthesis , Animals , Arthritis, Experimental/complications , Cell Line, Tumor , Corticotropin-Releasing Hormone/biosynthesis , Humans , Hypercalcemia/etiology , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Male , Neoplasms, Experimental/complications , Nerve Tissue Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Orexins , Pro-Opiomelanocortin/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Nude , Rats, Wistar
6.
Cancer Sci ; 102(6): 1208-15, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21401803

ABSTRACT

CD47 belongs to the immunoglobulin superfamily and is associated with Ɵ-integrins. Recently it was reported that CD47 ligation rapidly induces apoptosis in B-chronic lymphocytic leukemia (CLL) cells. Chronic lymphocytic leukemia is still an incurable hematological malignancy even with the novel therapeutic agents; therefore, new and effective agents for the treatment of CLL in clinical settings are urgently needed. We generated a murine monoclonal antibody against an extracellular domain of human CD47 (designated MABL). Subsequently, we created a disulfide-stabilized dimer of a single-chain antibody fragment of MABL (S-S diabody) to get rid of the adverse effect of MABL such as hemagglutination. In this study, we analyzed the effects of this new antibody on cellular proliferation, and the molecular mechanism of CD47-mediated apoptosis in human lymphoid malignant cells. Treatment with S-S diabody alone induced apoptosis of CD47-positive primary B-CLL and leukemic cells (MOLT-4 and JOK-1). In addition, administration of S-S diabody significantly prolonged the survival of severe combined immunodeficiency (SCID) mice inoculated with JOK-1 cells. In gene expression profiling of the S-S diabody-treated MOLT-4 cells, hypoxia inducible factor (HIF)-1α downstream genes (RTP801 and BNIP3) were upregulated, and the mRNA expression levels of HIF-1α, RTP801 and BNIP3 were increased. Knockdown of HIF-1α by siRNA repressed S-S diabody-induced apoptosis in MOLT4 cells. In conclusion, CD47 will be a molecular target for the treatment of lymphoid malignancies, and S-S diabody might have potential as a novel therapeutic agent for B-CLL.


Subject(s)
Apoptosis , CD47 Antigen/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Single-Chain Antibodies/immunology , Single-Chain Antibodies/therapeutic use , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Membrane Potentials , Membrane Proteins/genetics , Mice , Mice, SCID , Microscopy, Electron , Mitochondrial Proteins/genetics , Protein Multimerization , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Transcription Factors/genetics
7.
Am J Pathol ; 176(2): 968-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20035059

ABSTRACT

We investigated the roles of interleukin-6 (IL-6) and parathyroid hormone-related peptide (PTHrP) in oral squamous cell carcinoma (OSCC)-induced osteoclast formation. Microarray analyses performed on 43 human OSCC specimens revealed that many of the specimens overexpressed PTHrP mRNA, but a few overexpressed IL-6 mRNA. Immunohistochemical analysis revealed that IL-6 was expressed not only in cancer cells but also in fibroblasts and osteoclasts at the tumor-bone interface. Many of the IL-6-positive cells coexpressed vimentin. Conditioned medium (CM) derived from the culture of oral cancer cell lines (BHY, Ca9-22, HSC3, and HO1-u-1) stimulated Rankl expression in stromal cells and osteoclast formation. Antibodies against both human PTHrP and mouse IL-6 receptor suppressed Rankl in ST2 cells and osteoclast formation induced by CM from BHY and Ca9-22, although the inhibitory effects of IL6 antibody were greater than those of PTHrP antibody. CM derived from all of the OSCC cell lines effectively induced IL-6 expression in stromal cells, and the induction was partially blocked by anti-PTHrP antibody. Xenografts of HSC3 cells onto the periosteal region of the parietal bone in athymic mice presented histology and expression profiles of RANKL and IL-6 similar to those observed in bone-invasive human OSCC specimens. These results indicate that OSCC provides a suitable microenvironment for osteoclast formation not only by producing IL-6 and PTHrP but also by stimulating stromal cells to synthesize IL-6.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Cell Differentiation/genetics , Interleukin-6/physiology , Mouth Neoplasms/physiopathology , Osteoclasts/physiology , Parathyroid Hormone-Related Protein/physiology , Stromal Cells/metabolism , Adult , Aged , Aged, 80 and over , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Differentiation/drug effects , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-6/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Osteoclasts/metabolism , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone-Related Protein/pharmacology , Stromal Cells/drug effects , Stromal Cells/pathology , Transplantation, Heterologous
8.
J Neurooncol ; 103(2): 267-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20845060

ABSTRACT

To identify molecular therapeutic targets for glioma, we performed gene expression profiling by using a complementary DNA (cDNA) microarray method and identified the urokinase plasminogen activator receptor-associated protein (uPARAP/Endo180) as a gene expressed highly in glioma tissue compared with the normal brain tissue. The uPARAP is an endocytic receptor for collagen. In certain cell types, uPARAP occurs in a complex with the urokinase plasminogen activator receptor (uPAR) where it fulfills other functions in addition to collagenolysis. Quantitative PCR analysis using a cDNA panel revealed higher expression levels of uPARAP in glioma tissue compared with normal brain tissue. Western blot analysis revealed that the uPARAP protein was expressed in glioma samples and two glioma cell lines, KNS42 and KNS81, but not expressed in control tissue from the normal brain. Introduction of small interfering RNA-targeted uPARAP into the two different glioma cell lines, KNS42 and KNS81, resulted in downregulation of uPARAP expression, and it significantly suppressed glioma cell migration and invasion in vitro. Control glioma cells showed small cell bodies, whereas uPARAP siRNA-treated glioma cells exhibited large and flat morphology. Most of the polymeric actin in the control glioma cells was concentrated in the lamellipodia that are observed in mobile cells. In contrast, in the uPARAP siRNA-treated glioma cells, polymeric actin became organized in stress fibers and the lamellipodia disappeared, characteristic of immobile cells. Our present study suggests that uPARAP may be involved in glioma cell invasiveness through actin cytoskeletal rearrangement. downregulation of uPARAP may be a novel anti-invasion therapeutic strategy for malignant gliomas.


Subject(s)
Cell Movement/physiology , Cytoskeleton/metabolism , Glioma/metabolism , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Acetylcysteine , Blotting, Western , Cell Line, Tumor , Cytoskeleton/pathology , Down-Regulation , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Glioma/genetics , Glioma/pathology , Humans , Mannose-Binding Lectins/genetics , Membrane Glycoproteins/genetics , Neoplasm Invasiveness/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Receptors, Cell Surface/genetics
9.
Bioorg Med Chem Lett ; 21(6): 1795-801, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21316218

ABSTRACT

The MAP kinase pathway is one of the most important pathways involved in cell proliferation and differentiation, and its components are promising targets for antitumor drugs. Design and synthesis of a novel MEK inhibitor, based on the 3D-structural information of the target enzyme, and then multidimensional optimization including metabolic stability, physicochemical properties and safety profiles were effectively performed and led to the identification of a clinical candidate for an orally available potent MEK inhibitor, CH4987655, possessing a unique 3-oxo-oxazinane ring structure at the 5-position of the benzamide core structure. CH4987655 exhibits slow dissociation from the MEK enzyme, remarkable in vivo antitumor efficacy both in mono- and combination therapy, desirable metabolic stability, and insignificant MEK inhibition in mouse brain, implying few CNS-related side effects in human. An excellent PK profile and clear target inhibition in PBMC were demonstrated in a healthy volunteer clinical study.


Subject(s)
Antineoplastic Agents/chemistry , Benzamides/chemistry , MAP Kinase Kinase Kinases/antagonists & inhibitors , Oxazines/chemistry , Protein Kinase Inhibitors/chemistry , Administration, Oral , Allosteric Regulation , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacology , Humans , Models, Molecular , Oxazines/administration & dosage , Oxazines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology
10.
Cancer Sci ; 101(10): 2227-33, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20701608

ABSTRACT

The humanized monoclonal antibody (mAb) against CD317 antigen (anti-HM1.24 antibody; AHM), which is highly expressed on multiple myeloma (MM), induces antibody-dependent cellular cytotoxicity (ADCC). However, the antitumor activity of AHM in the clinical setting has not been clearly demonstrated. In this study, we produced defucosylated AHM and evaluated its potency for clinical application by performing autologous ADCC assays against primary MM cells from patients. Defucosylated AHM that was produced in rat myeloma YB2/0 cells expressing a low level of fucosyltransferase (FUT8) showed significant ADCC activity against three out of six primary MM cells in the presence of autologous PBMC, whereas conventional AHM did not. The results indicate that the potency of AHM to induce ADCC against primary MM cells was insufficient, but was significantly augmented by defucosylation. To generate more homogenous defucosylated monoclonal antibodies (mAb) for fermentation, we disrupted the GFT gene that encodes a GDP-fucose transporter in a CHO/DXB11 cell line by sequential homologous recombination. Analysis of the N-linked oligosaccharide in the defucosylated AHM produced by the established GFT(-/-)CHO cell line showed that a majority (93.4%) of the oligosaccharide was fucose free. The GFT(-/-) cells stably produced defucosylated mAb over passages. These results demonstrate that GTF(-/-)CHO-produced defucosylated AHM (GFTKO-AHM) will be a promising new therapeutic antibody against MM in the clinical setting.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Antigens, CD/immunology , Membrane Glycoproteins/immunology , Multiple Myeloma/drug therapy , Animals , CHO Cells , Cricetinae , Cricetulus , GPI-Linked Proteins , Glucosyltransferases/physiology , Humans , Monosaccharide Transport Proteins/physiology , Multiple Myeloma/immunology
11.
Anticancer Drugs ; 21(10): 907-16, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20847643

ABSTRACT

Glypican 3 (GPC3), a glycosylphosphatidylinositol-anchored heparan sulfate proteoglycan, is expressed in a majority of hepatocellular carcinoma tissues. The murine monoclonal antibody GC33 that specifically binds to the COOH-terminal part of GPC3 causes strong antibody-dependent cellular cytotoxicity against hepatocellular carcinoma cells and exhibits strong antitumor activity in the xenograft models. To apply GC33 for clinical use, we generated a humanized GC33 from complementarity-determining region grafting with the aid of both the hybrid variable region and two-step design methods. The humanized antibody bound to GPC3 specifically and induced antibody-dependent cellular cytotoxicity as effectively as a chimeric GC33 antibody. To improve stability of the humanized GC33, we further optimized humanized GC33 by replacing the amino acid residues that may affect the structure of the variable region of a heavy chain. Substitution of Glu6 with Gln in the heavy chain significantly improved the stability under high temperatures. GC33 also has the risk of deamidation of the -Asn-Gly- sequence in the complementarity-determining region 1 of the light chain. As substitution of Asn diminished the antigen binding, we changed the neighboring Gly to Arg to avoid deamidation. The resulting humanized anti-GPC3 antibody was as efficacious as chimeric GC33 against the HepG2 xenograft and is now being evaluated in clinical trials.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Glypicans/immunology , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Carcinoma, Hepatocellular/pathology , Complementarity Determining Regions/immunology , Drug Design , Humans , Immunoglobulin Variable Region/immunology , Liver Neoplasms/pathology , Mice , Protein Stability , Xenograft Model Antitumor Assays
12.
PLoS Negl Trop Dis ; 14(1): e0007860, 2020 01.
Article in English | MEDLINE | ID: mdl-31999695

ABSTRACT

In the past two decades there has been a significant expansion in the number of new therapeutic monoclonal antibodies (mAbs) that are approved by regulators. The discovery of these new medicines has been driven primarily by new approaches in inflammatory diseases and oncology, especially in immuno-oncology. Other recent successes have included new antibodies for use in viral diseases, including HIV. The perception of very high costs associated with mAbs has led to the assumption that they play no role in prophylaxis for diseases of poverty. However, improvements in antibody-expression yields and manufacturing processes indicate this is a cost-effective option for providing protection from many types of infection that should be revisited. Recent technology developments also indicate that several months of protection could be achieved with a single dose. Moreover, new methods in B cell sorting now enable the systematic identification of high-quality antibodies from humanized mice, or patients. This Review discusses the potential for passive immunization against schistosomiasis, fungal infections, dengue, and other neglected diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neglected Diseases/drug therapy , Animals , Dengue/drug therapy , Drug Development , Humans , Immunization, Passive , Mice , Mycoses/drug therapy , Schistosomiasis/drug therapy , Tropical Medicine
13.
Biochem Biophys Res Commun ; 378(2): 279-84, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19022220

ABSTRACT

Glypican 3 (GPC3), a GPI-anchored heparan sulfate proteoglycan, is expressed in the majority of hepatocellular carcinoma (HCC) tissues. Using MRL/lpr mice, we successfully generated a series of anti-GPC3 monoclonal antibodies (mAbs). GPC3 was partially cleaved between Arg358 and Ser359, generating a C-terminal 30-kDa fragment and an N-terminal 40-kDa fragment. All mAbs that induced antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC) against cells expressing GPC3 recognized the 30-kDa fragment, indicating that the C-terminal region of GPC3 serves as an epitope for mAb with ADCC and/or CDC inducing activities. Chimeric mAbs with Fc replaced by human IgG1 were created from GC33, one of the mAbs that reacted with the C-terminal 30-kDa fragment. Chimeric GC33 induced not only ADCC against GPC3-positive human HCC cells but also was efficacious against the Huh-7 human HCC xenograft. Thus, mAbs against the C-terminal 30-kDa fragment such as GC33 are useful in therapy targeting HCC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Carcinoma, Hepatocellular/drug therapy , Glypicans/antagonists & inhibitors , Liver Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , CHO Cells , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Glypicans/immunology , Humans , Immunodominant Epitopes/immunology , Liver Neoplasms/immunology , Mice , Neoplasm Proteins/immunology , Xenograft Model Antitumor Assays
14.
Oncol Rep ; 21(3): 747-55, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212635

ABSTRACT

We compared the antitumor efficacy and estrogen receptor (ER) degradation of CH4893237, a new orally active selective ER downregulator, with fulvestrant and tamoxifen in human breast cancer xenografts with low levels of serum estrogen (E2) (50.6, 22.9 and <16.7 pg/ml), equivalent to the ranges in postmenopausal or aromatase inhibitor-treated breast cancer patients. In addition, using proteolysis assays, we tested the conformational changes induced in ERalpha and ERbeta by CH4893237, fulvestrant, and 4-OH tamoxifen (4OHT). In ZR-75-1 xenografts with 50.6 pg/ml E2, CH4893237 (100 and 300 mg/kg/day p.o.) as well as fulvestrant (1 and 3 mg/body/week s.c.) showed complete growth inhibition (>90%) and tamoxifen (30 and 100 mg/kg/day p.o.) showed moderate tamoxifen resistance. The antitumor activity of CH4893237 (300 mg/kg) was the same as that of fulvestrant (3 mg/body) but the rate of ER degradation induced by CH4893237 (300 mg/kg) was significantly stronger than that of fulvestrant (3 mg/body) (94.3 vs. 85.5%, P<0.01). In Br-10 xenografts with 22.9 pg/ml E2, CH4893237 (30 mg/kg) and fulvestrant (1 mg/body) showed potent growth inhibition (>70%) whereas tamoxifen (1, 10 and 100 mg/kg) showed strong tamoxifen resistance. In Br-10 xenografts with ovariectomized-level E2 (<16.7 pg/ml), tamoxifen (30 mg/kg) increased the tumor volume but CH4893237 (30 mg/kg) showed no agonistic activity. In the ERalpha and ERbeta proteolysis assays, the band pattern for CH4893237 was different from fulvestrant. Thus, CH48793237 showed potent antitumor efficacies without agonistic activity and superior ER degradation in human breast cancer xenografts with low serum E2. Furthermore, the proteolysis studies suggest that CH4893237 induces conformational changes of ER different from those induced by fulvestrant. Therefore, CH4893237 alone or in combination with an aromatase inhibitor may be an efficient treatment for postmenopausal breast cancer patients.


Subject(s)
Breast Neoplasms/drug therapy , Estradiol/analogs & derivatives , Estrogens/blood , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Estradiol/pharmacology , Female , Fulvestrant , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
15.
Bioorg Med Chem Lett ; 19(7): 2018-21, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19254843

ABSTRACT

Novel hexacyclic camptothecin analogs containing cyclic amidine, urea, or thiourea moiety were designed and synthesized based on the proposed 3D-structure of the topoisomerase I (Topo I)/DNA/camptothecin ternary complex. The analogs were prepared from 9-nitrocamptothecin via 7,9-diaminocamptothecin derivatives as a key intermediate. Among them, 7c exhibited in vivo antitumor activities superior to CPT-11 in human cancer xenograft models in mice at their maximum tolerated doses though its in vitro antiproliferative activity was comparable to SN-38 against corresponding cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Camptothecin/analogs & derivatives , Topoisomerase I Inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Camptothecin/chemical synthesis , Camptothecin/chemistry , Camptothecin/pharmacology , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Humans , Mice , Structure-Activity Relationship , Transplantation, Heterologous
16.
Bioorg Med Chem Lett ; 19(10): 2772-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19362835

ABSTRACT

CH0793076 (1) is a novel hexacyclic camptothecin analog showing potent antitumor activity in various human caner xenograft models. To improve the water solubility of 1, water-soluble prodrugs were designed to generate an active drug 1 nonenzymatically, thus expected to show less interpatient PK variability than CPT-11. Among the prodrugs synthesized, 4c (TP300, hydrochloride) having a glycylsarcosyl ester at the C-20 position of 1 is highly water-soluble (>10mg/ml), stable below pH 4 and rapidly generates 1 at physiological pH in vitro. The rapid (ca. <1min) generation of 1 after incubation of TP300 with plasma (mouse, rat, dog and monkey) was also demonstrated. TP300 showed a broader antitumor spectrum and more potent antitumor activity than CPT-11 in various human cancer xenograft models.


Subject(s)
Antineoplastic Agents/chemical synthesis , Camptothecin/analogs & derivatives , Prodrugs/chemical synthesis , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Camptothecin/blood , Camptothecin/chemical synthesis , Camptothecin/chemistry , Camptothecin/pharmacokinetics , DNA Topoisomerases, Type I/metabolism , Dogs , Haplorhini , Humans , Hydrogen-Ion Concentration , Irinotecan , Mice , Mice, Nude , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats , Topoisomerase I Inhibitors , Transplantation, Heterologous , Water/chemistry
17.
Cancer Res ; 67(3): 1184-92, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17283154

ABSTRACT

Cross-linked human leukocyte antigen (HLA) class I molecules have been shown to mediate cell death in neoplastic lymphoid cells. However, clinical application of an anti-HLA class I antibody is limited by possible side effects due to widespread expression of HLA class I molecules in normal tissues. To reduce the unwanted Fc-mediated functions of the therapeutic antibody, we have developed a recombinant single-chain Fv diabody (2D7-DB) specific to the alpha2 domain of HLA-A. Here, we show that 2D7-DB specifically induces multiple myeloma cell death in the bone marrow environment. Both multiple myeloma cell lines and primary multiple myeloma cells expressed HLA-A at higher levels than normal myeloid cells, lymphocytes, or hematopoietic stem cells. 2D7-DB rapidly induced Rho activation and robust actin aggregation that led to caspase-independent death in multiple myeloma cells. This cell death was completely blocked by Rho GTPase inhibitors, suggesting that Rho-induced actin aggregation is crucial for mediating multiple myeloma cell death. Conversely, 2D7-DB neither triggered Rho-mediated actin aggregation nor induced cell death in normal bone marrow cells despite the expression of HLA-A. Treatment with IFNs, melphalan, or bortezomib enhanced multiple myeloma cell death induced by 2D7-DB. Furthermore, administration of 2D7-DB resulted in significant tumor regression in a xenograft model of human multiple myeloma. These results indicate that 2D7-DB acts on multiple myeloma cells differently from other bone marrow cells and thus provide the basis for a novel HLA class I-targeting therapy against multiple myeloma.


Subject(s)
HLA-A Antigens/immunology , Immunization, Passive/methods , Immunoglobulin Fragments/pharmacology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Death/immunology , Cell Line, Tumor , Cytokines/pharmacology , Enzyme Activation , HLA-A Antigens/biosynthesis , Humans , Jurkat Cells , Mice , Mice, SCID , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays , rho GTP-Binding Proteins/metabolism
18.
Int J Cancer ; 123(5): 1126-32, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18546273

ABSTRACT

Renal cell carcinomas (RCCs) are morphologically and genetically heterogeneous tumors and present diverse clinical courses. We developed a scoring system using levels of gene expression to predict the outcome for clear cell RCC patients. We selected differentially expressed genes from the DNA microarray data of 27 clear cell RCCs; 16 were metastasis phenotypes and 11 were not. We compared the selected gene set with previously published data and identified 33 overlapping genes closely associated with patient outcome. We selected the 12 top-ranked genes and confirmed the level of expression using quantitative reverse transcriptase PCR. Multivariate Cox analysis revealed that 3 genes-vascular cell adhesion molecule 1 (VCAM1), endothelin receptor type B (EDNRB), and regulator of G-protein signaling 5 (RGS5)-were the most tightly associated with cancer-specific survival and that higher expression of the 3 genes correlated with better outcome. A formula for an outcome predictor was generated from integration of the measurements of the expression levels of the 3 genes. Multivariate Cox models combined with a split-sample cross-validation method in a cohort of 386 clear cell RCC patients demonstrated that the derived score for outcome prediction was an independent predictor in cancer-specific survival tests. The accuracy of the prediction of cancer death after nephrectomy was improved by the inclusion of this score in receiver operating characteristic analysis from multivariate logistic regression models, suggesting that a scoring system based on the expression levels of these 3 genes is useful in the prediction of survival for patients with clear cell RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Gene Expression Profiling , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Adult , Aged , Carcinoma, Renal Cell/mortality , Cation Transport Proteins/genetics , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/mortality , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics
19.
Cancer Sci ; 99(12): 2461-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19032371

ABSTRACT

A murine (mAHM) and a humanized (AHM) monoclonal antibody against CD317 (also called tetherin, BST2, or HM1.24 antigen), expressed preferentially in neoplastic B cells such as multiple myeloma, exhibited antitumor effects as a result of antibody-dependent cellular cytotoxicity (ADCC). The putative interferon (IFN) response elements IRF-1/2 and ISGF3 are present in the promoter of the CD317 gene, and IFN has been used for the treatment of not only myeloproliferative diseases but also solid tumors such as renal cell carcinoma (RCC) and melanoma. Therefore, we examined the effects of IFN on the expression of CD317 and on the antitumor activity of AHM and mAHM in RCC and melanoma. Flow cytometry and in vitro ADCC assays with human or mouse effector cells demonstrated that IFN-alpha markedly increased the amount of cell surface CD317 and augmented the ADCC activity of mAHM and AHM in RCC cells and to a lesser extent in melanoma cells. Administration of IFN-alpha to mice bearing RCC xenografts also increased the expression of CD317 in tumor cells. When coadministered with IFN-alpha, mAHM exhibited more profound antitumor activity in both IFN-alpha-sensitive and -insensitive RCC xenograft models. Thus, AHM in combination with IFN-alpha may be an effective therapy for the treatment of RCC.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antigens, CD/immunology , Carcinoma, Renal Cell/therapy , Interferon-alpha/pharmacology , Kidney Neoplasms/therapy , Membrane Glycoproteins/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/genetics , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , GPI-Linked Proteins , Humans , Immunohistochemistry , Interferon-alpha/immunology , Kidney Neoplasms/immunology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Mice, SCID , Tumor Burden , Xenograft Model Antitumor Assays
20.
Oncol Rep ; 19(2): 489-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202799

ABSTRACT

We previously developed a DNA microarray-based system that out-performs traditionally used clinical parameters for prediction of early intrahepatic recurrence (IHR) of hepatocellular carcinoma (HCC). Because DNA microarray is too expensive for daily clinical use, we used a quantitative real-time reverse transcription-polymerase chain reaction (QRT-PCR) to develop a lower-cost predictor for early IHR. From the 12 early IHR-related genes integrated in the previous predictor, we selected 6 genes whose levels showed the strongest association between data from the 2 distinct DNA microarray platforms with the same sample set. Expression of these 6 genes relative to that of GAPDH was measured by QRT-PCR in 82 HCCs. Of the 82 HCCs, 39 and 43 were assigned to training and independent test sets, respectively. By searching all combinations (n=2-6) of the 6 genes, we found an optimal combination of 3 genes (HLADRA, DDX17 and LAPTM5) that minimized the leave-one-out error for prediction of early IHR in the training set. The 3-gene predictor constructed with the Fisher linear classifier correctly predicted early IHR or non-recurrence in 35 (81.4%) of 43 HCCs in the independent test set and had a high positive predictive value of 72.7% and a high negative predictive value of 84.4%. Multivariate analysis with the stepwise logistic regression showed that the 3-gene predictor [F(x)<0] was an independent risk factor for early IHR (risk ratio, 13.6; p=0.006), indicating its potential as an easy-to-use predictor for accurate prediction of early IHR of HCC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/surgery , Genes, Neoplasm , Liver Neoplasms/surgery , Neoplasm Recurrence, Local/diagnosis , Reverse Transcriptase Polymerase Chain Reaction/methods , Aged , DEAD-box RNA Helicases/genetics , Early Diagnosis , Female , Gene Expression , HLA-DR Antigens/genetics , Hepatectomy , Humans , Male , Membrane Proteins/genetics , Middle Aged , Prognosis , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL