Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
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
2.
Mol Cancer Ther ; 19(11): 2288-2297, 2020 11.
Article in English | MEDLINE | ID: mdl-32943545

ABSTRACT

Osimertinib is the only EGFR-tyrosine kinase inhibitor (TKI) capable of overcoming EGFR-T790M-mutated NSCLC, but osimertinib-resistant EGFR triple mutations (Del19/T790M/C797S or L858R/T790M/C797S) have been reported. Although allosteric EGFR TKIs (e.g., EAI-045) that potentially overcome L858R/T790M/C797S have been identified, there are no effective inhibitors against Del19/T790M/C797S. In this study, we identified CH7233163 as having the potential to overcome EGFR-Del19/T790M/C797S. CH7233163 showed potent antitumor activities against tumor with EGFR-Del19/T790M/C797S in vitro and in vivo In addition to EGFR-Del19/T790M/C797S, the characterization assays showed that CH7233163 more selectively inhibits various types of EGFR mutants (e.g., L858R/T790M/C797S, L858R/T790M, Del19/T790M, Del19, and L858R) over wild type. Furthermore, crystal structure analysis suggested that CH7233163 is a noncovalent ATP-competitive inhibitor for EGFR-Del19/T790M/C797S that utilizes multiple interactions with the EGFR's αC-helix-in conformation to achieve potent inhibitory activity and mutant selectivity. Therefore, we conclude that CH7233163 is a potentially effective therapy for osimertinib-resistant patients, especially in cases of EGFR-Del19/T790M/C797S.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Drug Resistance, Neoplasm/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Alleles , Amino Acid Substitution , Animals , Binding, Competitive , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Profiling , Humans , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/chemistry , Sequence Deletion , Structure-Activity Relationship , Xenograft Model Antitumor Assays
3.
Cancer Res ; 79(22): 5734-5745, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31391186

ABSTRACT

Targeting genetic alterations of oncogenes by molecular-targeted agents (MTA) is an effective approach for treating cancer. However, there are still no clinical MTA options for many cancers, including esophageal cancer. We used a short hairpin RNA library to screen for a new oncogene in the esophageal cancer cell line KYSE70 and identified YES proto-oncogene 1 (YES1) as having a significant impact on tumor growth. An analysis of clinical samples showed that YES1 gene amplification existed not only in esophageal cancer but also in lung, head and neck, bladder, and other cancers, indicating that YES1 would be an attractive target for a cancer drug. Because there is no effective YES1 inhibitor so far, we generated a YES1 kinase inhibitor, CH6953755. YES1 kinase inhibition by CH6953755 led to antitumor activity against YES1-amplified cancers in vitro and in vivo. Yes-associated protein 1 (YAP1) played a role downstream of YES1 and contributed to the growth of YES1-amplified cancers. YES1 regulated YAP1 transcription activity by controlling its nuclear translocation and serine phosphorylation. These findings indicate that the regulation of YAP1 by YES1 plays an important role in YES1-amplified cancers and that CH6953755 has therapeutic potential in such cancers. SIGNIFICANCE: These findings identify the SRC family kinase YES1 as a targetable oncogene in esophageal cancer and describe a new inhibitor for YES1 that has potential for clinical utility.See related commentary by Rai, p. 5702.


Subject(s)
Gene Amplification , Oncogenes , Cell Line, Tumor , Genes, src , Proto-Oncogene Proteins c-yes
4.
Mol Cancer Ther ; 17(12): 2519-2529, 2018 12.
Article in English | MEDLINE | ID: mdl-30242093

ABSTRACT

Members of the tropomyosin receptor kinase (TRK) family are expressed in their constitutively activated forms as a result of a gene fusion that occurs across a wide variety of cancer types. We have identified CH7057288 as a potent and selective TRK inhibitor that belongs to a novel chemical class. CH7057288 showed selective inhibitory activity against TRKA, TRKB, and TRKC in cell-free kinase assays and suppressed proliferation of TRK fusion-positive cell lines, but not that of TRK-negative cell lines. Strong in vivo tumor growth inhibition was observed in subcutaneously implanted xenograft tumor models of TRK fusion-positive cells. Furthermore, in an intracranial implantation model mimicking brain metastasis, CH7057288 significantly induced tumor regression and improved event-free survival. Recently, resistant mutations in the kinase domain of TRK have been reported in patients who show disease progression after treatment with the TRK inhibitors now under clinical development. Our compound maintained similar levels of in vitro and in vivo activity against one of these resistant mutants as it did to wild-type TRK. An X-ray crystal structure of the TRKA and CH7057288 complex supported the activity against the mutant. In addition, gene expression analysis revealed that CH7057288 suppressed MAPK and E2F pathways as downstream signaling of TRK fusion. Therefore, CH7057288 could be a promising therapeutic agent for TRK fusion-positive cancer.


Subject(s)
Benzofurans/pharmacology , Neoplasms/pathology , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , Protein Kinases/chemistry , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Cancer Lett ; 409: 116-124, 2017 11 28.
Article in English | MEDLINE | ID: mdl-28923400

ABSTRACT

Microphthalmia-associated transcription factor (MITF) is expressed in melanomas and has a critical role in melanocyte development and transformation. Because inhibition of MITF inhibits cell growth in melanoma, MITF is a potential therapeutic target molecule. Here, we report the identification of CH6868398, which has a novel chemical structure and suppresses MITF expression at the protein level in melanoma cells. CH6868398 showed cell growth inhibition activity against MITF-dependent melanoma cells both with and without BRAF mutation and also exhibited anti-tumor efficacy in a melanoma xenograft model. Because selective BRAF inhibitors are standard therapeutics for BRAF-mutated melanoma, we investigated the effect of CH6868398 with a BRAF inhibitor, PLX4720, on cell growth inhibition. The addition of CH6868398 enhanced the cell growth inhibition activity of PLX4720 in melanoma cell lines. Furthermore, combination of CH6868398 and PLX4720 efficiently suppressed MITF protein and enhanced cleavage of Caspase3 and poly (ADP-ribose) polymerase (PARP) in melanoma cell lines. These data support the therapeutic potential of CH6868398 as an anti-melanoma agent that reduces MITF protein levels in combination with BRAF inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Melanoma/drug therapy , Microphthalmia-Associated Transcription Factor/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , Indoles/pharmacology , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/administration & dosage , Random Allocation , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 17(10): 3272-81, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21558396

ABSTRACT

PURPOSE: The phosphatidylinositol 3-kinase (PI3K) pathway plays a central role in cell proliferation and survival in human cancer. PIK3CA mutations, which are found in many cancer patients, activate the PI3K pathway, resulting in cancer development and progression. We previously identified CH5132799 as a novel PI3K inhibitor. Thus, this study aimed to clarify the biochemical and antitumor activity of CH5132799 and elucidate the correlation between CH5132799 response and genetic alterations in the PI3K pathway. EXPERIMENTAL DESIGN: Kinase inhibitory activity was profiled in cell-free assays. A large panel of human breast, ovarian, prostate, and endometrial cancer cell lines, as well as xenograft models, were used to evaluate the antitumor activity of CH5132799, followed by analysis for genetic alterations. Effects on Akt phosphorylation induced by mTORC1 inhibition were tested with CH5132799 and compared with mTORC1 and PI3K/mTOR inhibitors. RESULTS: CH5132799 selectively inhibited class I PI3Ks and PI3Kα mutants in in vitro kinase assays. Tumors harboring PIK3CA mutations were significantly sensitive to CH5132799 in vitro and were remarkably regressed by CH5132799 in in vivo mouse xenograft models. In combination with trastuzumab, tumors disappeared in the trastuzumab-insensitive breast cancer model with the PIK3CA mutation. Moreover, CH5132799 did not reverse a negative feedback loop of PI3K/Akt/mTOR signaling and induced regression against tumors regrown after long-term mTORC1 inhibitor treatment. CONCLUSIONS: CH5132799 is a selective class I PI3K inhibitor with potent antitumor activity against tumors harboring the PIK3CA mutations. Prediction of CH5132799 response on the basis of PIK3CA mutations could enable patient stratification in clinical settings.


Subject(s)
Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Mutation/physiology , Neoplasms/genetics , Oncogenes/genetics , Oncogenes/physiology , Phosphatidylinositol 3-Kinases/metabolism , Substrate Specificity , Xenograft Model Antitumor Assays
7.
Cancer Chemother Pharmacol ; 65(2): 363-71, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19495753

ABSTRACT

PURPOSE: Identification of a novel topoisomerase I inhibitor which shows superior efficacy and less individual variation than irinotecan hydrochloride (CPT-11). METHODS: A novel camptothecin analog that is effective against breast cancer resistance protein (BCRP)-positive cells was screened, and a water soluble prodrug was generated. Antitumor activity of the prodrug was examined in BCRP-positive and -negative xenografts both as a single agent and in combination with other anti-cancer drugs. RESULTS: A novel camptothecin analog, CH0793076, was discovered. Because CH0793076 was found to be highly lipophilic, a water soluble prodrug (TP300) was generated. TP300 is stable in an acidic solution but is rapidly converted to CH0793076 under physiological pH conditions such as in sera. This efficient prodrug activation would minimize interpatient differences in pharmacokinetic and toxicity profiles. Unlike CPT-11, TP300 does not exhibit cholinergic interaction or cause acute diarrhea at effective doses. In mouse xenograft models, TP300 showed antitumor activity against both BCRP-positive and -negative xenografts, whereas CPT-11 was less active against BCRP-positive xenografts. In addition, the effective dose range (MTD/ED(50)) for TP300 was wider than for CPT-11 and TP300 showed additive or synergistic antitumor effects in combination with other anti-cancer drugs such as capecitabine, oxaliplatin, cisplatin, bevacizumab and cetuximab. CONCLUSION: It is therefore expected that TP300 will provide an additional treatment option for patients who will undergo chemotherapy with camptothecins.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Dipeptides/therapeutic use , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Neoplasm Proteins/biosynthesis , Prodrugs/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Acetylcholinesterase/metabolism , Animals , Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Dipeptides/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Irinotecan , Male , Mice , Mice, Nude , Prodrugs/pharmacology , Solubility , Water , Xenograft Model Antitumor Assays
8.
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
9.
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
10.
Int J Cancer ; 106(5): 799-805, 2003 Sep 20.
Article in English | MEDLINE | ID: mdl-12866042

ABSTRACT

Capecitabine is an orally available fluoropyrimidine and is finally converted to 5-FU selectively in tumor tissues. In our study, we examined whether the antitumor activity of capecitabine is directly affected by a modulation of dihydropyrimidine dehydrogenase (DPD). The modulations were carried out by the overexpression of DPD in tumor cells and by tumor selective DPD inhibition. The DPD-overexpressing cells were obtained by transfection of human DPD cDNA into HCT116 human colorectal cancer cells. The HCT116 cells bearing DPD cDNA expressed about 13 times higher DPD activities than the parental HCT116 cells, and they became significantly less susceptible to capecitabine than the parental cells when transplanted into nude mice. Administration of RO0094889 that is converted to a DPD inhibitor 5-vinyluracil selectively in tumor tissues restored the antitumor activity of capecitabine against the tumor of the HCT116 cells carrying DPD cDNA and various tumors expressing DPD. As compared to 5-ethynyluracil or 5-vinyluracil, which inhibited DPD not only in tumor tissues but also in other non-cancerous tissues, the effective dose range of RO0094889 in augmenting the efficacy of capecitabine was much broader. These results indicate that the antitumor activity of capecitabine is directly affected by DPD activities in tumor tissues and therefore, the combination of capecitabine and a tumor selective DPD inhibitor, RO0094889, will be beneficial to patients who have tumors with high levels of DPD.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colorectal Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Enzyme Inhibitors/therapeutic use , Oxidoreductases/antagonists & inhibitors , Animals , Blotting, Western , Capecitabine , Cell Division , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/prevention & control , DNA Primers/chemistry , DNA, Neoplasm/metabolism , Dihydrouracil Dehydrogenase (NADP) , Drug Synergism , Fluorouracil/analogs & derivatives , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured/transplantation , Xenograft Model Antitumor Assays
11.
Bioorg Med Chem Lett ; 13(5): 867-72, 2003 Mar 10.
Article in English | MEDLINE | ID: mdl-12617910

ABSTRACT

A series of tumor-activated prodrugs of the inhibitors of dihydropyrimidine dehydrogenase (DPD), an enzyme catabolizing 5-fluorouracil (5-FU: 4g), has been designed and synthesized. RO0094889 (11c) is a prodrug of 5-vinyluracil (4c), a known DPD inhibitor, and was designed to generate 4c selectively in tumor tissues by sequential conversion of 11c by three enzymes: esterase, cytidine deaminase and thymidine phosphorylase, the latter two of which are known to be highly expressed in various tumor tissues. When capecitabine (1), a tumor-activated prodrug of 5-FU, was co-administered orally with 11c, 5-FU in tumor tissues was significantly increased with only a slight increase of 5-FU in plasma as compared with oral capecitabine alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/chemical synthesis , Deoxycytidine/pharmacokinetics , Oxidoreductases/antagonists & inhibitors , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Uracil/analogs & derivatives , Administration, Oral , Animals , Capecitabine , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cytidine Deaminase/metabolism , Dihydrouracil Dehydrogenase (NADP) , Drug Design , Drug Stability , Esterases/metabolism , Female , Fluorouracil/blood , Fluorouracil/pharmacokinetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Prodrugs/administration & dosage , Thymidine Phosphorylase/metabolism , Tissue Distribution , Uracil/pharmacokinetics , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...