9 results on '"Survivin inhibitor"'
Search Results
2. Phase I safety and pharmacokinetic study of YM155, a potent selective survivin inhibitor, in combination with erlotinib in patients with EGFR TKI refractory advanced non-small cell lung cancer.
- Author
-
Shimizu, Toshio, Nishio, Kazuto, Sakai, Kazuko, Okamoto, Isamu, Okamoto, Kunio, Takeda, Masayuki, Morishita, Maiko, and Nakagawa, Kazuhiko
- Subjects
- *
NON-small-cell lung carcinoma , *PEMETREXED , *CLINICAL trial registries , *PHARMACOKINETICS , *THERAPEUTIC use of antineoplastic agents , *LUNG cancer , *QUINONE , *RESEARCH , *PROTEIN kinase inhibitors , *RESEARCH methodology , *LUNG tumors , *CELL receptors , *PROGNOSIS , *EVALUATION research , *MEDICAL cooperation , *IMIDAZOLES , *COMPARATIVE studies , *RESEARCH funding , *DRUG resistance in cancer cells , *LONGITUDINAL method , *PHARMACODYNAMICS - Abstract
Purpose: This phase I study was conducted to evaluate the safety and pharmacokinetics of YM155, a potent, selective survivin inhibitor, in combination with erlotinib in patients with EGFR TKI refractory advanced non-small cell lung cancer (NSCLC).Methods: The pimary objectives were to evaluate the safety and tolerability of YM155 at escalating doses (3.6, 4.8, 6.0, and 8.0 mg/m2/days) administered every 3 weeks as continuous intravenous infusion over 168 h in combination with erlotinib at a fixed dose (150 mg, once a day). Secondary objectives were to assess the pharmacokinetics of YM155, antitumor activity, and the relationship between biomarkers and efficacy. The changes in survivin expression in biopsied tumor pre- and post-YM155 administration and serum cytokine levels were also analyzed.Results: Fifteen patients were treated. The most common YM155-related adverse event was the presence of urine microalbumin, whereas grades 3/4 toxicities were rare. One patient who received 4.8 mg/m2/days YM155 developed a dose-limiting grade 2 serum creatinine elevation. YM155 exposure in plasma showed dose proportionality across all dose ranges tested. No pharmacokinetic interaction occurred between YM155 and erlotinib. The serum cytokines IL-8, G-CSF, and MIP-1b showed decreasing trends in patients who achieved progression-free survival of ≥ 12 weeks. Durable stable disease for ≥ 24 weeks was observed in two patients.Conclusion: Up to 8.0 mg/m2/days YM155 administered every 3 weeks in combination with erlotinib exhibited a favorable safety profile and moderate clinical efficacy. These results suggest that inhibiting survivin is a potential therapeutic strategy for select patients with EGFR TKI refractory NSCLC.Trial Registration: UMIN000031912 at UMIN Clinical Trials Registry (UMIN-CTR). [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
3. Synthesis and antitumor activity evaluation of asiatic acid derivatives as survivin inhibitor.
- Author
-
Meng, Yan-Qiu, Cui, Hua-Bo, Li, Lei, Zhang, Wei-Chen, Pan, Hong-Shuang, Yu, Ting-Ting, and Li, Wei
- Subjects
- *
PACLITAXEL , *ANTINEOPLASTIC agents , *CELL lines , *CELL migration , *DRUG interactions , *HYDROCARBONS , *MICROBIOLOGICAL assay , *MOLECULAR structure , *PROTEINS , *TERPENES , *CHEMICAL inhibitors - Abstract
A series of asiatic acid derivatives were synthesized and their cytotoxicities in vitro against two cancer cell lines (HepG2 and SGC7901) were evaluated by MTT assay. The results showed that compounds I2, I6, and II6 have more potent anticancer activity than that of the positive control drug paclitaxel. The interactions between the compounds I2, I6, and II6 and survivin were also studied by docking simulations. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
4. Synthesis and biological evaluation of novel 2-arylvinyl-substituted naphtho[2,3-d]imidazolium halide derivatives as potent antitumor agents.
- Author
-
Wei, Qingyun, Li, Ju, Tang, Feng, Yin, Yin, Zhao, Yong, and Yao, Qizheng
- Subjects
- *
NAPHTHOL derivatives , *CHEMICAL synthesis , *IMIDAZOLES , *ANTINEOPLASTIC agent synthesis , *BROMIDES , *VINYLATION , *APOPTOSIS , *WESTERN immunoblotting , *CASPASES - Abstract
Two series of novel 2-arylvinyl-naphtho[2,3- d ]imidazol-3-ium iodide derivatives and 2-arylvinyl-naphtho[2,3- d ]imidazol-3-ium bromide derivatives were designed and synthesized by the structural combination of YM155 with stilbenoids. All compounds were tested for anti-proliferative activity against PC-3, A375 and HeLa human cancer cell lines. Two of the compounds were selected for further investigation: 12b, which showed potent cytotoxicity against the three tested cell lines with IC 50 values in the range of 0.06–0.21 μM, and 7l , which displayed excellent selectivity for PC-3 cells with an IC 50 of only 22 nM. Western blot analysis results indicated that both 12b and 7l suppress the expression of Bcl-2 and Survivin proteins, which helps induce apoptosis. As determined by the percent of Annexin V-FITC-positive apoptotic cells, 12b was not only significantly more effective than 7l at a concentration of 100 nM in PC-3 cells but also induced apoptosis in a dose-dependent manner with more potency than 7l at a concentration of 1000 nM in A375 cells. Therefore, compound 12b was chosen for further in-depth studies investigating the mechanism of apoptosis. The results showed that it could activate caspase-3, hydrolyze PARP, and even inactivate ERK. Moreover, 12b arrested A375 cells at S phase in a time-dependent and dose-dependent manner, while having a visible effect on microtubule dynamics. In addition, ( E )-2-(2-(1 H -indol-3-yl)vinyl)-1-benzyl-3-(2-methoxyethyl)-4,9-dioxo-4,9-dihydro-1 H -naphtho[2,3-d]imidazol-3-ium bromide ( 12b ) exhibited significant antitumor activity when evaluated in a subcutaneous solid tumor model. Our study reveals that 2-arylvinyl-substituted naphtho[2,3-d]imidazolium scaffolding is a promising new entity for the development of multi-target anticancer drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
5. Preventing Pluripotent Cell Teratoma in Regenerative Medicine Applied to Hematology Disorders
- Author
-
Benoit Rucheton, François Béliveau, Aurélie Bedel, Véronique Guyonnet-Duperat, Hubert de Verneuil, Isabelle Moranvillier, Sandrine Dabernat, Benoit Rousseau, Isabelle Lamrissi-Garcia, François Moreau-Gaudry, and Bruno Cardinaud
- Subjects
0301 basic medicine ,Time Factors ,Survivin ,Cellular differentiation ,Mice, SCID ,Regenerative Medicine ,Hematopoietic stem cell ,0302 clinical medicine ,Translational Research Articles and Reviews ,Mice, Inbred NOD ,Induced pluripotent stem cell ,Genes, Transgenic, Suicide ,Hematopoietic Stem Cell Transplantation ,Imidazoles ,Teratoma ,Hematology ,General Medicine ,Caspase 9 ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Haematopoiesis ,Cell Transformation, Neoplastic ,Phenotype ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Safety ,Stem cell ,Pluripotent Stem Cells ,Induced Pluripotent Stem Cells ,Survivin inhibitor ,Biology ,Risk Assessment ,Tacrolimus ,Cell Line ,iCaspase‐9 ,03 medical and health sciences ,medicine ,Animals ,Humans ,Cell Proliferation ,Dose-Response Relationship, Drug ,Cell Biology ,Suicide gene ,Hematopoietic Stem Cells ,medicine.disease ,Hematologic Diseases ,Xenograft Model Antitumor Assays ,Thymidine kinase ,030104 developmental biology ,Cancer research ,Naphthoquinones ,Developmental Biology - Abstract
Iatrogenic tumorigenesis is a major limitation for the use of human induced pluripotent stem cells (hiPSCs) in hematology. The teratoma risk comes from the persistence of hiPSCs in differentiated cell populations. Our goal was to evaluate the best system to purge residual hiPSCs before graft without compromising hematopoietic repopulation capability. Teratoma risk after systemic injection of hiPSCs expressing the reporter gene luciferase was assessed for the first time. Teratoma formation in immune-deficient mice was tracked by in vivo bioimaging. We observed that systemic injection of hiPSCs produced multisite teratoma as soon as 5 weeks after injection. To eliminate hiPSCs before grafting, we tested the embryonic-specific expression of suicide genes under the control of the pmiR-302/367 promoter. This promoter was highly active in hiPSCs but not in differentiated cells. The gene/prodrug inducible Caspase-9 (iCaspase-9)/AP20187 was more efficient and rapid than thymidine kinase/ganciclovir, fully specific, and without bystander effect. We observed that iCaspase-9-expressing hiPSCs died in a dose-dependent manner with AP20187, without reaching full eradication in vitro. Unexpectedly, nonspecific toxicity of AP20187 on iCaspase-9-negative hiPSCs and on CD34+ cells was evidenced in vitro. This toxic effect strongly impaired CD34+-derived human hematopoiesis in adoptive transfers. Survivin inhibition is an alternative to the suicide gene approach because hiPSCs fully rely on survivin for survival. Survivin inhibitor YM155 was more efficient than AP20187/iCaspase-9 for killing hiPSCs, without toxicity on CD34+ cells, in vitro and in adoptive transfers. hiPSC purge by survivin inhibitor fully eradicated teratoma formation in immune-deficient mice. This will be useful to improve the safety management for hiPSC-based medicine.
- Published
- 2016
- Full Text
- View/download PDF
6. Inhibition of skeletal growth of human prostate cancer by the combination of docetaxel and BKM1644: an aminobisphosphonate derivative
- Author
-
Zhengjia Chen, Xin Li, Lajos Gera, Daqing Wu, Omer Kucuk, Shumin M. Zhang, and Kenza Mamouni
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_treatment ,Docetaxel ,urologic and male genital diseases ,Mice ,Random Allocation ,Prostate cancer ,0302 clinical medicine ,Drug Interactions ,docetaxel resistance ,Neoplasm Metastasis ,STAT3 ,bone metastasis ,survivin inhibitor ,Diphosphonates ,biology ,Bone metastasis ,prostate cancer ,preclinical models ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,Taxoids ,Research Paper ,medicine.drug ,medicine.medical_specialty ,Mice, Nude ,Antineoplastic Agents ,Bone Neoplasms ,03 medical and health sciences ,Cell Line, Tumor ,Survivin ,medicine ,Animals ,Humans ,neoplasms ,Cell Proliferation ,business.industry ,Cell growth ,Prostatic Neoplasms ,Bisphosphonate ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,STAT protein ,Cancer research ,biology.protein ,business - Abstract
Bone metastasis is a major cause of prostate cancer (PCa) morbidity and mortality. Despite some success in transiently controlling clinical symptoms with docetaxel-based therapy, PCa patients become docetaxel-resistant and inevitably progress with no cure. We synthesized an acyl-tyrosine bisphosphonate amide derivative, BKM1644, with the intent of targeting bone metastatic PCa and enhancing docetaxel's efficacy. BKM1644 exhibits potent anti-cancer activity in the NCI-60 panel and effectively inhibits the proliferation of metastatic, castration-resistant PCa (mCRPC) cells, with IC50 ranging between 2.1 μM and 6.3 μM. Significantly, BKM1644 sensitizes mCRPC cells to docetaxel treatment. Mice with pre-established C4-2 tumors in the tibia show a marked decrease in serum prostate-specific antigen (control: 173.72 ± 37.52 ng/ml, combined treatment: 64.45 ± 22.19 ng/ml; p < 0.0001) and much improved bone architecture after treatment with the combined regimen. Mechanistic studies found that docetaxel temporarily but significantly increases survivin, an anti-apoptotic protein whose overexpression has been correlated with PCa bone metastasis and therapeutic resistance. Intriguingly, BKM1644 effectively inhibits survivin expression, which may antagonize docetaxel-induced survivin in bone metastatic PCa cells. Signal transducer and activator of transcription 3 (Stat3) may be involved in the suppression of survivin transcription by BKM1644, as confirmed by a survivin reporter assay. Collectively, these data indicate that BKM1644 could be a promising small-molecule agent to improve docetaxel efficacy and retard the bone metastatic growth of PCa.
- Published
- 2016
- Full Text
- View/download PDF
7. Discovery of Novel Tubulin Inhibitors and Selective Survivin Inhibitors for Advanced Melanoma and Total Synthesis of Bioactive 20S-hydroxyvitamin D3
- Author
-
Wang, Qinghui
- Subjects
- medicinal chemistry, microtubule inhibitor, survivin inhibitor, synthetic chemistry, total synthesis, vitamin D3, Diseases, Medicinal and Pharmaceutical Chemistry, Medicine and Health Sciences, Neoplasms, Pharmaceutics and Drug Design, Pharmacy and Pharmaceutical Sciences
- Abstract
According to the statistics from American Cancer Society, the 5-year survival rate for patients with advanced melanoma is as low as 5%. Treatment of advanced melanoma, therefore, represents an unmet medical need. In this dissertation, I will show the effort to develop new generations of bioavailable tubulin inhibitors targeting the colchicine binding site and selective small-molecule survivin inhibitors for treating advanced melanoma. Extensive structure-activity relationship (SAR) studies of lead molecules ABI-231 and UC-112 have been performed. Chapter 1 will introduce the current situation of advanced or metastatic melanoma, its clinical drug treatments, as well as problems in current drug treatments. Microtubule dynamics and survivin will be discussed as promising therapeutic targets for developing anticancer drugs. 20S-hydroxyvitamin D3 (20S-OH-D3) will be introduced as a promising anti-inflammatory scaffold. Chapter 2 will disclose the SAR study of ABI-231, a previously reported potent tubulin inhibitor from our lab. In this chapter, a new synthetic method was developed to enable the synthesis of ABI-231 analogues modifying the indole moiety. The novel synthetic method involved the synthesis of a key diamine intermediate and imidazoline formation. From the new synthetic method, thirty ABI-231 analogues were synthesized and tested for activities. Among all analogues, 10ab with a 4-methyl-3-indole moiety and 10bb with a 4-indole moiety showed the most potent antiproliferative activities against a panel of melanoma cell lines. 10ab and 10bb had IC50s of 2.2 and 3.0 nM, respectively. The SAR result revealed that modification of the indole moiety in ABI-231 was beneficial to activity. In Chapter 3, we will describe our effort to develop the SAR study of ABI-231 focusing on modification of the 3,4,5-TMP moiety. This is selected since it is one of the most common moieties in current tubulin inhibitors targeting the colchicine binding site. To circumvent the use of potentially explosive azide reported in Chapter 2, an alternative was established to efficiently generate ABI-231 analogues. This new synthetic method involved Suzuki coupling and Grignard reactions to modify the 3,4,5-TMP moiety and to produce target compounds in gram-scale. Among the eight analogues synthesized, the one containing an unique 3-methoxybenzo[4,5]-dioxene moiety had the strongest antiproliferative activity against a panel of melanoma cell lines with an average IC50 of 1.9 nM. To our best knowledge, it represents the most successful instance of isosterically modifying the 3,4,5-TMP moiety in CA-4 derivatives. Chapter 4 will highlight our effort to synthesize reverse ABI (RABI) analogues for SAR study. In this chapter, a novel and concise synthetic route was established toaccess RABI scaffold. RABI scaffold was constructed through a Grignard reaction/Suzuki-Miyaura coupling reaction strategy. From this new synthetic method, twelve novel RABI analogues were synthesized. Compared to MX-RABI (the previously reported most potent RABI), several new RABI analogues showed significantly improved cytotoxicities. In particular, analogue 15i with a 4-indazole moiety showed the most potent antiproliferative activity against a panel of melanoma cell lines and had an average IC50 of 0.8 nM. This is the first sub-nM anti-tubulin compound in the related scaffolds. Chapter 5 will reveal our latest SAR study of UC-112, a previously reported selective survivin inhibitor. Fourteen UC-112 analogues modifying the benzyloxy moiety of UC-112 were synthesized. Their corresponding SAR result demonstrated that indole moiety was the most favorable (analogue 12a). Subsequent structural optimization of 12a by introducing either mono-substituent or di-substituent to the indole moiety led to the synthesis of another twenty-four new UC-112 analogues. Several substituted indole analogues showed equipotency to that of UC-112 and MX-106. Importantly, new indole analogues exhibited significant abilities to overcome multidrug-resistance mediated by Pgp overexpression. Chapter 6 is characterized by the establishment of a total synthetic method of 20SOH-D3 which showed comparable antiproliferative activity to 1,25α-dihydroxyvitamin D3 without hypercalcemic toxic effect upto a concentration of 60 μg/kg in vivo. The total synthesis of 20S-OH-D3 involved parallel generation of key intermediates from ergocalciferol. The vitamin D3 core structure was constructed through Wittig-Horner coupling reaction. Deprotection of SEM and TBS was achieved in one step. 20S-OH-D3 was furnished in sixteen steps with an overall yield of 0.4%.
- Published
- 2017
8. Overcoming Acquired Resistance to BRAF Inhibitors by Novel Synergistic Drug Combination and Discovery of Novel Smac Mimetics as Selective Survivin Inhibitors
- Author
-
Wang, Jin
- Subjects
- BRAF inhibitor, combination therapy, inhibitor of apoptosis, melanoma, survivin inhibitor, tubulin inhibitor, Medicinal and Pharmaceutical Chemistry, Medicine and Health Sciences, Pharmaceutics and Drug Design, Pharmacy and Pharmaceutical Sciences
- Abstract
The first part (Chapter 1 and 2) of this dissertation presents a novel combination study of melanoma therapy. Acquired clinical resistance to vemurafenib, a selective BRAFV600E inhibitor, arises frequently after short term chemotherapy. Since the inhibitions of targets in the RAFMEK-ERK pathway result in G0/G1 cell cycle arrest, vemurafenib-resistant cancer cells are expected to escape this cell cycle arrest and progress to subsequent G2/M phase. We hypothesized that a combined therapy using vemurafenib with a G2/M phase blocking agent will trap resistant cells and overcome vemurafenib resistance. To test this hypothesis, we first determined the combination index (CI) values of our novel tubulin inhibitor ABI-274 and vemurafenib on parental human A375 and MDA-MB-435 melanoma cell lines to be 0.32 and 0.1, respectively, suggesting strong synergy for the combination. We then developed an A375RF21 subline with significant acquired resistance to vemurafenib and confirmed the strong synergistic effect. Next we studied the potential mechanisms of overcoming vemurafenib resistance. Flow cytometry confirmed that the combination of ABI-274 and vemurafenib synergistically arrested cells in G1/G2/M phase, and significantly increased apoptosis in both parental A375 and the vemurafenib-resistant A375RF21 cells. Western blot analysis revealed that the combination treatment effectively reduced the level of phosphorylated and total AKT, activated the apoptosis cascade, and increased cleaved caspase-3 and cleaved PARP, but had no significant influence on the level of ERK phosphorylation. Finally, in vivo co-administration of vemurafenib with ABI-274 showed strong synergistic efficacy in the vemurafenib-resistant xenograft model in nude mice. Overall, these results offer a rational combination strategy to significantly enhance the therapeutic benefit in melanoma patients who inevitably become resistant to current BRAF inhibition therapy. The second part (Chapter 3 to 5) of this dissertation focuses on the discovery of a series of small molecule survivin inhibitors. Inhibitors of apoptosis (IAP) proteins are widely considered as promising cancer drug targets, especially for drug-resistant tumors. Mimicking the IAP-binding motif of second mitochondria-derived activator of caspases (Smac) is a rational strategy to design potential IAP inhibitors. In this report, we used the bioactive conformation of AVPI tetrapeptide in the N-terminus of Smac as a template and performed a shape-based virtual screening against a drug-like compound library to identify novel IAP inhibitors. Top hits were subsequently docked to available IAP crystal structures as a secondary screening followed by validation using in vitro biological assays. Four novel hit compounds were identified that potently inhibited cell growth in two human melanoma (A375 and M14) and two human prostate (PC-3 and DU145) cancer cell lines. The best compound, UC-112, has IC50 values ranging from 0.7 to 3.4 µM. UC-112 also potently inhibits the growth of P-glycoprotein (Pgp) overexpressed multidrug-resistant cancer cells, strongly activates capase-3/7 and caspase-9 activities, and selectively down-regulates survivin level at a concentration as low as 1 µM. Co-incubation of UC-112 with a known proteasome inhibitor (MG132) rescued survivin inhibition, consistent with the anticipated mechanism of action for UC-112. As a single agent, UC-112 strongly inhibits tumor growth and reduces both XIAP and survivin levels in an A375 human melanoma xenograft model in vivo. Three analogs generated from UC-112 structural modification along with template compound UC-112 were submitted to NCI-60 cancer cell line screening. The results indicated that structural modification of UC-112 to give our best compound MX106 has improved activity by four fold (2.2 µM for UC-112 vs. 0.5 µM for MX106, average GI50 values over all cancer cell lines in the NCI-60 panel).Western blot analyses demonstrated the new compounds maintained high selectivity for survivin inhibition over other members in the inhibitiors of apoptosis protein family. When tested in an A375 human melanoma xenograft model, the most active compound MX106 effectively suppressed tumor growth and strongly induced cancer cell apoptosis in tumor tissues. Taken together, this novel scaffold is promising for the development of selective survivin inhibitors as potential anticancer agents.
- Published
- 2015
9. Anticancer drug FL118 is more than a survivin inhibitor: where is the Achilles' heel of cancer?
- Author
-
Li F
- Abstract
Can a solution be found that overcomes all chemotherapy and/or radiation resistance resulting from different genetic and epigenetic alternations in various cancer types? The answer is likely NO. However, there are two ways that may be followed to approach this goal. One way is through the use of poly-therapies that target multiple mechanisms to kill cancer cells, which is the current state of the art. This approach raises issues of high costs and/or toxic limitations, since the toxicities of each agent are often additive. This poly-pharmacy approach has not proven to be a major success, although it has proven to be superior to most current mono-pharmacy approaches. The other way to approach the goal is to find a single anticancer drug that targets multiple different treatment resistant mechanisms. In this regard, a small chemical molecule (FL118) was recently discovered by serendipity during targeted discovery of anticancer drugs using the survivin gene as a target and biomarker. FL118 was found to not only inhibit multiple antiapoptotic proteins (survivin, XIAP, cIAP2) in the inhibitor of apoptosis (IAP) family, but to also inhibit the antiapoptotic protein Mcl-1 in the Bcl-2 family, while inducing the pro-apoptotic proteins Bax and Bim expression. Importantly, inhibition of these target genes and of tumor growth by FL118 is independent of p53 status (wild type, mutant or null), although mechanisms of action may be distinct among cells with different p53 status. Therefore, FL118 may effectively control cancer that loses functional p53, in which most DNA damage drugs (if not all) show a marked lack of efficiency. Recent studies further revealed that the superior anticancer activity of FL118 is highly dependent on its primary structure and steric configuration, suggesting that FL118 may be a promising drug platform for generating novel derivatives based on its core structure. Intriguingly, although FL118 has structural similarity to irinotecan and topotecan, two FDA-approved topoisomerase 1 (Top1) inhibitors for cancer treatment, cancer cells with Top1 mutations shows little contributions of treatment resistance to FL118 antitumor activity, while strikingly increasing irinotecan and topotecan resistance. Furthermore, both irinotecan and topotecan are the efflux pump ABCG2 substrates; cancer cells with high expression of ABCG2 showed strong irinotecan and topotecan resistance. In contrast, FL118 is not an ABCG2 substrate; ABCG2 overexpression in cancer cells does not show resistance to FL118 treatment. Current evidence suggests that future studies may unravel more unexpected mechanisms of action for this unique small molecule FL118.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.