Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Anticancer Res ; 36(11): 5937-5944, 2016 11.
Article in English | MEDLINE | ID: mdl-27793919

ABSTRACT

BACKGROUND: Multiple factors affect the in vivo antitumor activity of antibody-based therapeutics; however, the influence of cell surface retention on antibody-dependent cellular cytotoxicity (ADCC) is not fully understood. Here we evaluated the importance of cell surface antibody retention in antitumor activity mediated by ADCC in vivo. MATERIALS AND METHODS: Two mAbs against tumor-associated calcium signal transducer 2 (TACSTD2/TROP2), AR47A6.4.2 and Pr1E11, were used. Antitumor activities against BxPC3 and Colo205 cells were investigated through in vitro and in vivo assays. RESULTS: Pr1E11 showed better cell surface retention than AR47A6.4.2 in vitro although Pr1E11 and AR47A6.4.2 showed equivalent ADCC activity. Complement-dependent cytotoxicity and antiproliferative activity were not observed for either antibody. Pr1E11 exhibited higher antitumor activity than AR47A6.4.2 in vivo. CONCLUSION: Our results suggest that high cell surface retention can result in potent ADCC activity in vivo. This observation could provide novel insight into how effectively screen for antibodies with strong in vivo antitumor activity.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Humans , Xenograft Model Antitumor Assays
2.
Biochem Biophys Res Commun ; 458(4): 877-82, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25701778

ABSTRACT

TROP-2 is a type Ⅰ transmembrane glycoprotein that is highly expressed in various epithelial cancer cells, and its increased expression correlates with poor prognosis. Although several anti-TROP-2 antibodies have been described, they were found unsuitable for antitumor therapy use in vivo as naked antibodies. In this study, we established a novel anti-TROP-2 antibody, designated Pr1E11, from mice immunized with primary prostate cancer cells. Antibody screening was based on the infection activity of Adv-LacZ-FZ33, which displays an immunoglobulin G binding domain in the adenoviral fiber protein. We found that Pr1E11 specifically binds to TROP-2 with high affinity and recognizes diverse epithelial cancer cell lines and primary pancreatic cancer tissues. Epitope analysis using TROP-2 deletion mutants revealed that binding site of Pr1E11 is a cysteine-rich domain, a unique epitope compared with other available anti-TROP-2 antibodies. In addition, Pr1E11 exhibited low internalization activity, which may make it suitable for naked antibody therapeutics. Our results suggest that Pr1E11 may stimulate different biological activities from other anti-TROP-2 antibodies based on its unique binding epitope, and is a potential candidate for naked antibody therapeutics for various epithelial cancer treatments.


Subject(s)
Adenoviridae/immunology , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Epitopes/immunology , Prostatic Neoplasms/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Humans , Male , Mice, Inbred BALB C , Molecular Sequence Data , Prostate/immunology , Sequence Deletion , Tumor Cells, Cultured
3.
Anticancer Res ; 34(1): 89-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24403448

ABSTRACT

Prostate-specific membrane antigen (PSMA) is an attractive target for treatment of prostate cancer. Using the PSMA-recognizing mouse monoclonal antibody 2C9 obtained in our previous study, the biological activities of PSMA antibody were evaluated. Mouse-human chimeric IgG1 of 2C9 (KM2777) showed antibody-dependent cellular cytotoxicity activity against PSMA-expressing prostate cancer cells in the presence of human peripheral blood mononuclear cells (PBMCs). To increase lymphocyte-mediated cytotoxicity of KM2777, C-terminus interleukin-2 (IL-2)-fused KM2777 (KM2812) was constructed. KM2812 retained binding activity to PSMA and exhibited growth-stimulating activity equivalent to IL-2 on the IL-2-dependent T-cell line CTLL-2. Moreover, KM2812 exhibited enhanced cytotoxic activity against PSMA-expressing prostate cancer cells in the presence of PBMCs compared with KM2777. In a xenograft tumor model using PSMA-expressing prostate cancer cells, KM2812 exhibited marked antitumor activity, accompanied by complete regression of tumor in some of the KM2812-treated mice. These results suggest that KM2812 has a therapeutic potential for prostate cancer by stimulating lymphocyte-mediated antitumor cytotoxicity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Surface/immunology , Cytotoxicity, Immunologic/immunology , Glutamate Carboxypeptidase II/immunology , Interleukin-2/immunology , Membrane Glycoproteins/immunology , Prostatic Neoplasms/therapy , Animals , Antibody-Dependent Cell Cytotoxicity , Flow Cytometry , Glutamate Carboxypeptidase II/antagonists & inhibitors , Humans , Interleukin-2/metabolism , Leukocytes, Mononuclear , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, SCID , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Pharmacol Sci ; 123(3): 256-66, 2013.
Article in English | MEDLINE | ID: mdl-24162023

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is a Ca(2+)-permeable non-selective cation channel that transmits pain signals. TRPV1 is activated by multiple stimuli such as capsaicin, acid, and heat. During inflammation, TRPV1 is reported to be sensitized by protein kinase C (PKC) in dorsal root ganglia (DRG) neurons, which leads to reduction in the threshold of the temperature for TRPV1 activation to body temperature. This sensitization is considered to contribute to chronic inflammatory pain. In a previous study, we discovered orally active 5,5-diarylpentadienamide TRPV1 antagonists. To examine the effects of our TRPV1 antagonists on PKC-sensitized TRPV1, we developed an in vitro assay system to monitor the TRPV1 sensitization by PKC. In this assay system, our TRPV1 antagonists, such as (2E,4Z)-N-[(3R)-3-hydroxy-2-oxo-1,2,3,4-tetrahydro-5-quinolyl]-5-(4-isopropoxyphenyl)-5-(4-trifluoromethylphenyl)-2,4-pentadienamide (K-685), inhibited the activation of TRPV1 sensitized by PKC. The potentiation of heat-induced inward currents by PKC was seen in rat DRG neurons, and K-685 attenuated these currents. Furthermore, K-685 reversed the thermal hyperalgesia and mechanical allodynia in a rat complete Freund's adjuvant-induced inflammatory pain model. These results therefore suggest that K-685 has a strong potential as a new analgesic drug for the treatment of inflammatory pain.


Subject(s)
Analgesics , Chronic Pain/drug therapy , Chronic Pain/genetics , Freund's Adjuvant/adverse effects , Inflammation/complications , Pentanoic Acids/pharmacology , Pentanoic Acids/therapeutic use , Protein Kinase C/physiology , Quinolones/pharmacology , Quinolones/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Animals , Chronic Pain/etiology , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley
5.
J Med Chem ; 55(7): 3436-51, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22394104

ABSTRACT

We have developed a novel and potent chemical series of 5,5-diphenylpentadienamides for targeting TRPV1 in vitro and in vivo. In this investigation, we examined a variety of replacements for the 5-position of dienamides with the goal of addressing issues related to pharmacokinetics. Our data suggest that substitution with alkoxy groups on the phenyl ring at the 5-position increases their ability to penetrate the blood-brain barrier. This investigation culminated in the discovery of compound (R)-36b, which showed a good pharmacokinetic profile. In vivo, compound (R)-36b was found to be effective at reversing mechanical allodynia in rats in a dose-dependent manner, and it reversed thermal hyperalgesia in a model of neuropathic pain induced by sciatic nerve injury.


Subject(s)
Alkadienes/chemical synthesis , Amides/chemical synthesis , Analgesics/chemical synthesis , Quinolones/chemical synthesis , TRPV Cation Channels/antagonists & inhibitors , Alkadienes/pharmacokinetics , Alkadienes/pharmacology , Amides/pharmacokinetics , Amides/pharmacology , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Blood-Brain Barrier/metabolism , Calcium/metabolism , Capsaicin/pharmacology , Dogs , Epstein-Barr Virus Nuclear Antigens/genetics , HEK293 Cells , Haplorhini , Humans , Hyperalgesia/prevention & control , Male , Microsomes, Liver/metabolism , Neuralgia/prevention & control , Quinolones/pharmacokinetics , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics
6.
Life Sci ; 88(19-20): 898-907, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21466810

ABSTRACT

AIMS: Benidipine, a dihydropyridine Ca(2+) channel blocker, has been reported to block T-type Ca(2+) channels; however, the mechanism underlying this effect was unclear. In this study, we characterized the mechanism responsible for this blocking activity. Furthermore, the blocking activity was compared between two enantiomers of benidipine, (S, S)- and (R, R)-benidipine. MAIN METHODS: Human Ca(v)3.2 (hCa(v)3.2) T-type Ca(2+) channels stably expressed in the human embryonic kidney cell line, HEK-293, were studied in whole-cell patch-clamp recordings and Ca(2+) mobilization assay. KEY FINDINGS: In whole-cell patch-clamp recordings, benidipine blocked hCa(v)3.2 T-type Ca(2+) currents elicited by depolarization to a comparable extent as efonidipine. The block was dependent on stimulation frequency and holding potential, but not test potential. Benidipine significantly shifted the steady-state inactivation curve to the hyperpolarizing direction, but had no effect on the activation curve. Benidipine prolonged the recovery from inactivation of hCa(v)3.2 T-type Ca(2+) channels without any effect on the kinetics of activation, inactivation, or deactivation. In the Ca(2+) mobilization assay, benidipine was more potent than efonidipine in blocking Ca(2+) influx through hCa(v)3.2 T-type Ca(2+) channels. (S, S)-Benidipine was more potent than (R, R)-benidipine in blocking hCa(v)3.2 T-type Ca(2+) currents, but there was no difference in blocking the Ca(2+) influx. SIGNIFICANCE: We have characterized the blocking activity of benidipine against hCa(v)3.2 Ca(2+) channels and revealed the difference between the two enantiomers of benidipine. The blocking action of benidipine could be mediated by stabilizing hCa(v)3.2 Ca(2+) channels in an inactivated state.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/physiology , Dihydropyridines/pharmacology , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology
7.
Circ Res ; 91(10): 961-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12433842

ABSTRACT

In neonatal cardiomyocytes, activation of the G(q)-coupled alpha(1)-adrenergic receptor (alpha(1)AR) induces hypertrophy by activating mitogen-activated protein kinases, including c-Jun NH(2)-terminal kinase (JNK). Here, we show that JNK activation is essential for alpha(1)AR-induced hypertrophy, in that alpha(1)AR-induced hypertrophic responses, such as reorganization of the actin cytoskeleton and increased protein synthesis, could be blocked by expressing the JNK-binding domain of JNK-interacting protein-1, a specific inhibitor of JNK. We also identified the classes and subunits of G proteins that mediate alpha(1)AR-induced JNK activation and hypertrophic responses by generating several recombinant adenoviruses that express polypeptides capable of inhibiting the function of specific G-protein subunits. alpha(1)AR-induced JNK activation was inhibited by the expression of carboxyl terminal regions of Galpha(q), Galpha(12), and Galpha(13). JNK activation was also inhibited by the Galpha(q/11)- or Galpha(12/13)-specific regulator of G-protein signaling (RGS) domains and by C3 toxin but was not affected by treatment with pertussis toxin or by expression of the carboxyl terminal region of G protein-coupled receptor kinase 2, a polypeptide that sequesters Gbetagamma. alpha(1)AR-induced hypertrophic responses were inhibited by Galpha(q/11)- and Galpha(12/13)-specific RGS domains, C3 toxin, and the carboxyl terminal region of G protein-coupled receptor kinase 2 but not by pertussis toxin. Activation of Rho was inhibited by carboxyl terminal regions of Galpha(12) and Galpha(13) but not by Galpha(q). Our findings suggest that alpha(1)AR-induced hypertrophic responses are mediated in part by a Galpha(12/13)-Rho-JNK pathway, in part by a G(q/11)-JNK pathway that is Rho independent, and in part by a Gbetagamma pathway that is JNK independent.


Subject(s)
Adaptor Proteins, Signal Transducing , Cardiomegaly/metabolism , DNA-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Myocardium/metabolism , Receptors, Adrenergic, alpha-1/metabolism , ADP Ribose Transferases/pharmacology , Adenoviridae/genetics , Animals , Animals, Newborn , Botulinum Toxins/pharmacology , Cardiomegaly/etiology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cells, Cultured , DNA-Binding Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13 , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/genetics , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Oxidants/pharmacology , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Pertussis Toxin/pharmacology , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transfection , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
8.
J Pharmacol Exp Ther ; 301(1): 51-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11907156

ABSTRACT

(-)-1-(3,4-Dimethoxyphenetylamino)-3-(3,4-dihydroxy)-2-propanol [(-)-RO363] is a highly selective beta(1)-adrenergic receptor (beta(1)AR) agonist. To study the binding site of beta(1)-selective agonist, chimeric beta(1)/beta(2)ARs and Ala-substituted beta(1)ARs were constructed. Several key residues of beta(1)AR [Leu(110) and Thr(117) in transmembrane domain (TMD) 2], and Phe(359) in TMD 7] were found to be responsible for beta(1)-selective binding of (-)-RO363, as determined by competitive binding. Based on these results, we built a three-dimensional model of the binding domain for (-)-RO363. The model indicated that TMD 2 and TMD 7 of beta(1)AR form a binding pocket; the methoxyphenyl group of N-substituent of (-)-RO363 seems to locate within the cavity surrounded by Leu(110), Thr(117), and Phe(359). The amino acids Leu(110) and Phe(359) interact with the phenyl ring of (-)-RO363, whereas Thr(117) forms hydrogen bond with the methoxy group of (-)-RO363. To examine the interaction of these residues with beta(1)AR in an active state, each of the amino acids was changed to Ala in a constitutively active (CA)-beta(1)AR mutant. The degree of decrease in the affinity of CA-beta(1)AR for (-)-RO363 was essentially the same as that of wild-type beta(1)AR when mutated at Leu(110) and Thr(117). However, the affinity was decreased in Ala-substituted mutant of Phe(359) compared with that of wild-type beta(1)AR. These results indicated that Leu(110) and Thr(117) are necessary for the initial binding of (-)-RO363 with beta(1)-selectivity, and interaction of Phe(359) with the N-substituent of (-)-RO363 in an active state is stronger than in the resting state.


Subject(s)
Adrenergic beta-1 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Amino Acids/drug effects , Catechols/pharmacology , Propanolamines/pharmacology , Adrenergic beta-2 Receptor Agonists , Amino Acid Sequence , Gene Expression Regulation , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation/physiology , Plasmids/genetics , Radioligand Assay , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
9.
Biochem Biophys Res Commun ; 291(4): 995-1000, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11866464

ABSTRACT

In rat neonatal myocytes, a constitutively active G alpha(q) causes cellular injury and apoptosis. However, stimulation of the alpha(1)-adrenergic receptor, one of the G(q) protein-coupled receptors, with phenylephrine for 48 h causes little cellular injury and apoptosis. Expression of the G beta gamma-sequestering peptide beta ARK-ct increases the phenylephrine-induced cardiac injury, indicating that G beta gamma released from G(q) counteracts the G alpha(q)-mediated cellular injury. Stimulation with phenylephrine activates extracellular signal-regulated kinase (ERK) and Akt, and activation is significantly blunted by beta ARK-ct. Inhibition of Akt by inhibitors of phosphatidylinositol 3-kinase increases the cellular injury induced by phenylephrine stimulation. In contrast to the inhibition of Akt, inhibition of ERK does not affect the phenylephrine-induced cardiac injury. These results suggest that G beta gamma released from G(q) upon alpha(1)-adrenergic receptor stimulation activates ERK and Akt. However, activation of Akt but not ERK plays an important role in the protection against the G alpha(q)-induced cellular injury and apoptosis.


Subject(s)
GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Heterotrimeric GTP-Binding Proteins/metabolism , Myocardium/metabolism , Peptides , Protein Serine-Threonine Kinases , Receptors, Adrenergic, alpha-1/metabolism , Recombinant Proteins , Signal Transduction , Animals , Animals, Newborn , Apoptosis , Carrier Proteins/pharmacology , Cell Respiration/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/genetics , Kinetics , Mitochondria/drug effects , Mitochondria/physiology , Mitogen-Activated Protein Kinases/metabolism , Mutation , Myocardium/cytology , Phenylephrine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...