1
|
Huang M, Kang Z, Li S, Zhang B, Xiao Y, Li S, Li W. Phase I clinical trial of a novel procaspase-3 activator SM-1 with temozolomide in recurrent high-grade gliomas. Neoplasia 2025; 61:101141. [PMID: 39970841 PMCID: PMC11879598 DOI: 10.1016/j.neo.2025.101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
OBJECTIVE Despite a standard of care, the mortality of recurrent high-grade gliomas (HGGs) remains high. SM-1 is a novel molecular activator that has shown to target procaspase-3, which is overexpressed in HGGs. A phase I clinical trial was conducted to evaluate the safety, pharmacokinetics, and primary clinical efficacy of SM-1 plus TMZ. Participants received escalating doses of daily oral SM-1 (450, 600, and 800 mg) plus standard TMZ therapy. METHODS In the preclinical study, the synergistic effects of SM-1 and temozolomide (TMZ) in rodent models were evaluated. In the clinical study, adult patients received SM-1 therapy in various doses in combination with a standard TMZ dosing. The tolerability and pharmacokinetics data of the combination therapy were tested. The primary efficacy was measured by tumor response in accordance with the RANO criteria. RESULTS A total of 13 patients with recurrent HGG were enrolled, with 11 patients completed ≥ two cycles of therapy and received tumor assessment. Among them, one patient had complete response, whereas two patients had partial response for the best change from baseline. No dose-limited toxicities were observed, and no maximum tolerated dose was reached. CONCLUSION SM-1 has the potential to enhance antitumor activity while alleviating the side effects of TMZ. SM-1 exhibited mild toxicity in patients with recurrent HGG. The combination of SM-1 and TMZ warrants further investigation, with promising clinical outcomes. The monotherapy phase and expansion phase of SM-1 are still ongoing. (ClinicalTrials.gov number, CTR20221641).
Collapse
Affiliation(s)
- Mengqian Huang
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
| | - Zhuang Kang
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Shenglan Li
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Botao Zhang
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Yantao Xiao
- Clinical R&D Department, Shenzhen Zhenxing Pharmaceutical Technology Co., Ltd. , PR China.
| | - Shangwei Li
- Clinical R&D Department, Shenzhen Zhenxing Pharmaceutical Technology Co., Ltd. , PR China.
| | - Wenbin Li
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
2
|
Ai N, Yuan H, Liang Y, Lu S, Ouyang D, Lai QH, Lai LL. Multi-View Multiattention Graph Learning With Stack Deep Matrix Factorization for circRNA-Drug Sensitivity Association Identification. IEEE J Biomed Health Inform 2024; 28:7670-7682. [PMID: 39186430 DOI: 10.1109/jbhi.2024.3431693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Identifying circular RNA (circRNA)-drug sensitivity association (CDsA) is crucial for advancing drug development. As conducting traditional wet experiments for determining CDsA is costly and inefficient, calculation methods have already proven to be a valid approach to cope with this problem. However, there exists limited research addressing the prediction of the CDsA prediction problem, and certain discrepancies persist, particularly concerning false-negative associations. As a consequence, we present a multi-view framework, called MAGSDMF, for identifying latent CDsA. Firstly, MAGSDMF applies ultiple ttention mechanisms and raph learning methods to dynamically extract features and strengthen the features of inside and across multi-similarity networks of circRNA and drug. Secondly, the tack eep atrix Factorization (SDMF) is devised to directly extract features from CDsAs. We consider multi-similarity networks with the original CDsAs as multi-view information. Thirdly, MAGSDMF utilizes a multi-attention channel mechanism to integrate these features for the purpose of reconstructing CDsA. Finally, MAGSDMF performs another DMF based on the reconstruction to identify the latent CDsAs. Simultaneously, contrastive learning (CL) is implemented to enhance the generalization capability of MAGSDMF and oversee the learning process of the underlying links prediction task. In comparative experiments, MAGSDMF achieves superior performance on two datasets with AUC values of 0.9743 and 0.9739 based on 5-fold cross-validation. Moreover, in case studies, the achievements further validate the identification reliability of MAGSDMF.
Collapse
|
3
|
Bimoussa A, Oubella A, Alossaimi MA, Aziz M, Attaullah HM, Ejaz SA, Morjani H, Auhmani A, Robert A, Riahi A, Riadi Y, Ait Itto MY. Novel Bis-1,2,3-triazole-thiazolidinone hybrid as anticancer agents that induce apoptosis and molecular modeling study. Future Med Chem 2024; 16:2193-2210. [PMID: 39387360 PMCID: PMC11622789 DOI: 10.1080/17568919.2024.2394019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/12/2024] [Indexed: 10/15/2024] Open
Abstract
Aim: A series of (R)-Carvone-based 1,2,3-triazole-thiazolidinone 17a-h hybrids were synthesized and characterized by spectroscopic techniques NMR and HRMS. The chemical reactivity and the stability parameters were observed via DFT.Method/results: The objective was to evaluate the anticancer activity of the synthesized compounds against cancer cell lines. The mechanism of action by which the 17b and 17g exert their effect suggested that they may induce apoptosis through activation of caspase-3/7. This effect was observed against the most important NIMA-related kinases via Docking investigation. The designed compounds were identified as the best inhibitors of the NEK family via the inactivation of the caspase-3. The Docking results were supported by Dynamics where the binding energies justified the medicinal importance of the synthesized derivatives.
Collapse
Affiliation(s)
- Abdoullah Bimoussa
- Laboratory of Organic Synthesis & Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Université Cadi Ayyad, BP PO Box 2390, Marrakech, 40001, Morocco
| | - Ali Oubella
- Laboratory of Organic & Physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, Ibnou Zohr University, Agadir, Morocco
| | - Manal A Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Mubashir Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Hafiz Muhammad Attaullah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Hamid Morjani
- BioSpectroscopie Translationnelle, BioSpecT-EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51 Rue Cognacq Jay, 51096, Reims Cedex, France
| | - Aziz Auhmani
- Laboratory of Organic Synthesis & Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Université Cadi Ayyad, BP PO Box 2390, Marrakech, 40001, Morocco
| | - Anthony Robert
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire Université de Reims Champagne-Ardenne, Bat. Europol'Agro - Moulin de La Housse UFR SciencesB.P.,1039, 51687, Reims Cedex 2, France
| | - Abdelkhalek Riahi
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire Université de Reims Champagne-Ardenne, Bat. Europol'Agro - Moulin de La Housse UFR SciencesB.P.,1039, 51687, Reims Cedex 2, France
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Moulay Youssef Ait Itto
- Laboratory of Organic Synthesis & Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Université Cadi Ayyad, BP PO Box 2390, Marrakech, 40001, Morocco
| |
Collapse
|
4
|
Oubella A, Alossaimi MA, Riadi Y, Bhat MA, Bakheit AH, Taha ML, Auhmani A, Morjani H, Geesi MH, Ait Itto MY. Thiazolidinone-linked-1,2,3-triazoles with (R)-Carvone as new potential anticancer agents. Future Med Chem 2024; 16:1449-1464. [PMID: 39190475 DOI: 10.1080/17568919.2024.2351287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/25/2024] [Indexed: 08/28/2024] Open
Abstract
Aim: This study explores the cytotoxic and apoptotic effects of novel thiazolidinone-1,2,3-triazole hybrids on HT-1080, A-549, and MDA-MB-231 cancer cell lines.Methods & results: The synthesized compounds underwent comprehensive characterization (NMR and HRMS) to confirm their structures and purity. Subsequent anticancer activity screening across diverse cancer cell lines revealed promising antitumor potential notably, compounds 6f and 6g. Mechanistic investigations unveiled that compound 6f triggers apoptosis through the caspase-3/7 pathway. In terms of in silico studies, the compound 6f was identified as a potent inhibitor of caspase-3 and caspase-7.Conclusion: The present study underscores the therapeutic potential of thiazolidinone-1,2,3-triazole hybrids against certain cancer cells. These findings highlight a promising avenue for the development of cancer treatment strategies utilizing these (R)-Carvone-based derivatives.
Collapse
Affiliation(s)
- Ali Oubella
- Laboratory of Organic & Physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, Iboun Zohr University, Agadir, Morocco
| | - Manal A Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ahmed Hassan Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohamed Labd Taha
- Laboratory of Organic & Physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, Iboun Zohr University, Agadir, Morocco
| | - Aziz Auhmani
- Laboratory of Organic Synthesis & Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Université Cadi Ayyad, BP PO Box 2390, Marrakech, 40001, Morocco
| | - Hamid Morjani
- BioSpectroscopie Translationnelle, BioSpecT-EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51 Rue Cognacq Jay, Reims Cedex, 51096, France
| | - Mohammed H Geesi
- Department of Chemistry, College of Science & Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Moulay Youssef Ait Itto
- Laboratory of Organic Synthesis & Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Université Cadi Ayyad, BP PO Box 2390, Marrakech, 40001, Morocco
| |
Collapse
|
5
|
Lu S, Liang Y, Li L, Liao S, Zou Y, Yang C, Ouyang D. Inferring circRNA-drug sensitivity associations via dual hierarchical attention networks and multiple kernel fusion. BMC Genomics 2023; 24:796. [PMID: 38129810 PMCID: PMC10734204 DOI: 10.1186/s12864-023-09899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Increasing evidence has shown that the expression of circular RNAs (circRNAs) can affect the drug sensitivity of cells and significantly influence drug efficacy. Therefore, research into the relationships between circRNAs and drugs can be of great significance in increasing the comprehension of circRNAs function, as well as contributing to the discovery of new drugs and the repurposing of existing drugs. However, it is time-consuming and costly to validate the function of circRNA with traditional medical research methods. Therefore, the development of efficient and accurate computational models that can assist in discovering the potential interactions between circRNAs and drugs is urgently needed. In this study, a novel method is proposed, called DHANMKF , that aims to predict potential circRNA-drug sensitivity interactions for further biomedical screening and validation. Firstly, multimodal networks were constructed by DHANMKF using multiple sources of information on circRNAs and drugs. Secondly, comprehensive intra-type and inter-type node representations were learned using bi-typed multi-relational heterogeneous graphs, which are attention-based encoders utilizing a hierarchical process. Thirdly, the multi-kernel fusion method was used to fuse intra-type embedding and inter-type embedding. Finally, the Dual Laplacian Regularized Least Squares method (DLapRLS) was used to predict the potential circRNA-drug sensitivity associations using the combined kernel in circRNA and drug spaces. Compared with the other methods, DHANMKF obtained the highest AUC value on two datasets. Code is available at https://github.com/cuntjx/DHANMKF .
Collapse
Affiliation(s)
- Shanghui Lu
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
- School of Mathematics and Physics, Hechi University, No.42, Longjiang, 546300, Guangxi, China
| | - Yong Liang
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China.
- Peng Cheng Laboratory, Shenzhen, 518055, Guangdong, China.
| | - Le Li
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
| | - Shuilin Liao
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
| | - Yongfu Zou
- School of Mathematics and Physics, Hechi University, No.42, Longjiang, 546300, Guangxi, China
| | - Chengjun Yang
- School of Artificial Intelligence and Manufacturing, Hechi University, No.42, Longjiang, 546300, Guangxi, China
| | - Dong Ouyang
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
| |
Collapse
|
6
|
Boudreau MW, Tonogai EJ, Schane CP, Xi MX, Fischer JH, Vijayakumar J, Ji Y, Tarasow TM, Fan TM, Hergenrother PJ, Dudek AZ. The combination of PAC-1 and entrectinib for the treatment of metastatic uveal melanoma. Melanoma Res 2023; 33:514-524. [PMID: 37738028 PMCID: PMC10615773 DOI: 10.1097/cmr.0000000000000927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
The treatment of metastatic uveal melanoma remains a major clinical challenge. Procaspase-3, a proapoptotic protein and precursor to the key apoptotic executioner caspase-3, is overexpressed in a wide range of malignancies, and the drug PAC-1 leverages this overexpression to selectively kill cancer cells. Herein, we investigate the efficacy of PAC-1 against uveal melanoma cell lines and report the synergistic combination of PAC-1 and entrectinib. This preclinical activity, tolerability data in mice, and the known clinical effectiveness of these drugs in human cancer patients led to a small Phase 1b study in patients with metastatic uveal melanoma. The combination of PAC-1 and entrectinib was tolerated with no treatment-related grade ≥3 toxicities in these patients. The pharmacokinetics of entrectinib were not affected by PAC-1 treatment. In this small and heavily pretreated initial cohort, stable disease was observed in four out of six patients, with a median progression-free survival of 3.38 months (95% CI 1.6-6.5 months). This study is an initial demonstration that the combination of PAC-1 and entrectinib may warrant further clinical investigation. Clinical trial registration: Clinical Trials.gov: NCT04589832.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Emily J. Tonogai
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Claire P. Schane
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Min X. Xi
- HealthPartners Institute, Minneapolis, MN, USA
| | - James H. Fischer
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Yan Ji
- HealthPartners Institute, Minneapolis, MN, USA
| | | | - Timothy M. Fan
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Vanquish Oncology, Inc, Champaign, IL
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802 USA
| | - Paul J. Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Vanquish Oncology, Inc, Champaign, IL
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Arkadiusz Z. Dudek
- HealthPartners Institute, Minneapolis, MN, USA
- Vanquish Oncology, Inc, Champaign, IL
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
7
|
Testolin G, Richter J, Ritter A, Prochnow H, Köhnke J, Brönstrup M. Optical Modulation of Antibiotic Resistance by Photoswitchable Cystobactamids. Chemistry 2022; 28:e202201297. [PMID: 35771231 DOI: 10.1002/chem.202201297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 01/07/2023]
Abstract
The rise of antibiotic resistance causes a serious health care problem, and its counterfeit demands novel, innovative concepts. The combination of photopharmacology, enabling a light-controlled reversible modulation of drug activity, with antibiotic drug design has led to first photoswitchable antibiotic compounds derived from established scaffolds. In this study, we converted cystobactamids, gyrase-inhibiting natural products with an oligoaryl scaffold and highly potent antibacterial activities, into photoswitchable agents by inserting azobenzene in the N-terminal part and/or an acylhydrazone moiety near the C-terminus, yielding twenty analogs that contain mono- as well as double-switches. Antibiotic and gyrase inhibition properties could be modulated 3.4-fold and 5-fold by light, respectively. Notably, the sensitivity of photoswitchable cystobactamids towards two known resistance factors, the peptidase AlbD and the scavenger protein AlbA, was light-dependent. While irradiation of an analog with an N-terminal azobenzene with 365 nm light led to less degradation by AlbD, the AlbA-mediated inactivation was induced. This provides a proof-of-principle that resistance towards photoswitchable antibiotics can be optically controlled.
Collapse
Affiliation(s)
- Giambattista Testolin
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Jana Richter
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Antje Ritter
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Hans Prochnow
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Jesko Köhnke
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.,German Center for Infection Research (DZIF) Site Hannover-Braunschweig, 38124, Braunschweig, Germany.,Center of Biomolecular Drug Research (BMWZ), Leibniz University, 30159, Hannover, Germany
| |
Collapse
|
8
|
Yalçınkaya S, Yalçın Azarkan S, Karahan Çakmakçı AG. Determination of the effect of L. plantarum AB6-25, L. plantarum MK55 and S. boulardii T8-3C microorganisms on colon, cervix, and breast cancer cell lines: Molecular docking, and molecular dynamics study. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Deng L, Liu Z, Qian Y, Zhang J. Predicting circRNA-drug sensitivity associations via graph attention auto-encoder. BMC Bioinformatics 2022; 23:160. [PMID: 35508967 PMCID: PMC9066932 DOI: 10.1186/s12859-022-04694-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/20/2022] [Indexed: 11/18/2022] Open
Abstract
Background Circular RNAs (circRNAs) play essential roles in cancer development and therapy resistance. Many studies have shown that circRNA is closely related to human health. The expression of circRNAs also affects the sensitivity of cells to drugs, thereby significantly affecting the efficacy of drugs. However, traditional biological experiments are time-consuming and expensive to validate drug-related circRNAs. Therefore, it is an important and urgent task to develop an effective computational method for predicting unknown circRNA-drug associations. Results In this work, we propose a computational framework (GATECDA) based on graph attention auto-encoder to predict circRNA-drug sensitivity associations. In GATECDA, we leverage multiple databases, containing the sequences of host genes of circRNAs, the structure of drugs, and circRNA-drug sensitivity associations. Based on the data, GATECDA employs Graph attention auto-encoder (GATE) to extract the low-dimensional representation of circRNA/drug, effectively retaining critical information in sparse high-dimensional features and realizing the effective fusion of nodes’ neighborhood information. Experimental results indicate that GATECDA achieves an average AUC of 89.18% under 10-fold cross-validation. Case studies further show the excellent performance of GATECDA. Conclusions Many experimental results and case studies show that our proposed GATECDA method can effectively predict the circRNA-drug sensitivity associations.
Collapse
Affiliation(s)
- Lei Deng
- School of Software, Xinjiang University, Urumqi, China.,School of Computer Science and Engineering, Central South University, Changsha, China
| | - Zixuan Liu
- School of Software, Xinjiang University, Urumqi, China
| | - Yurong Qian
- School of Software, Xinjiang University, Urumqi, China
| | - Jingpu Zhang
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan, China.
| |
Collapse
|
10
|
Dung DTM, Park EJ, Anh DT, Phan DTP, Na IH, Kwon JH, Kang JS, Tung TT, Han SB, Nam NH. Design, synthesis and evaluation of novel 2-oxoindoline-based acetohydrazides as antitumor agents. Sci Rep 2022; 12:2886. [PMID: 35190616 PMCID: PMC8861050 DOI: 10.1038/s41598-022-06887-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 02/01/2022] [Indexed: 11/29/2022] Open
Abstract
In our search for novel small molecules activating procaspase-3, we have designed and synthesized two series of novel (E)-N'-arylidene-2-(2-oxoindolin-1-yl)acetohydrazides (4) and (Z)-2-(5-substituted-2-oxoindolin-1-yl)-N'-(2-oxoindolin-3-ylidene)acetohydrazides (5). Cytotoxic evaluation revealed that the compounds showed notable cytotoxicity toward three human cancer cell lines: colon cancer SW620, prostate cancer PC-3, and lung cancer NCI-H23. Especially, six compounds, including 4f–h and 4n–p, exhibited cytotoxicity equal or superior to positive control PAC-1, the first procaspase-3 activating compound. The most potent compound 4o was three- to five-fold more cytotoxic than PAC-1 in three cancer cell lines tested. Analysis of compounds effects on cell cycle and apoptosis demonstrated that the representative compounds 4f, 4h, 4n, 4o and 4p (especially 4o) accumulated U937 cells in S phase and substantially induced late cellular apoptosis. The results show that compound 4o would serve as a template for further design and development of novel anticancer agents.
Collapse
Affiliation(s)
- Do T M Dung
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Eun J Park
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Duong T Anh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Dung T P Phan
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Ik H Na
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Joo H Kwon
- Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, Republic of Korea
| | - Jong S Kang
- Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, Republic of Korea
| | - Truong T Tung
- Faculty of Pharmacy, PHENIKAA University, Hanoi, 12116, Vietnam.,PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi, 12116, Vietnam
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea.
| | - Nguyen-Hai Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam.
| |
Collapse
|
11
|
Oubella A, Fawzi M, Bimoussa A, N’Ait Ousidi A, Auhmani A, Riahi A, Robert A, El Firdoussi L, Morjani H, Ait Itto MY. Convenient route to benzo[1,2,3]selenadiazole–isoxazole hybrids and evaluation of their in vitro cytotoxicity. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02083-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
12
|
Dung DTM, Park EJ, Anh DT, Hai PT, Bao LQ, Ji AY, Kang JS, Tung TT, Han SB, Nam NH. Design, Synthesis and Evaluation of Novel (E)-N'-((1-(4-chlorobenzyl)-1H-indol-3-yl)methylene)-2-(4-oxoquinazolin-3(4H)-yl)acetohydrazides as Antitumor Agents. Anticancer Agents Med Chem 2022; 22:2586-2598. [PMID: 35040418 DOI: 10.2174/1871520622666220118154914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/21/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Herein, we have designed and synthesized a series of the novel (E)-N'-((1-(4-chlorobenzyl)-1H-indol-3-yl)methylene)-2-(4-oxoquinazolin-3(4H)-yl)acetohydrazides (5) as potent small molecules activating procaspase-3. The compounds were designed by the amalgamation of structural features of PAC-1 (the first procaspase-3 activator) and oncrasin-1, one potential anticancer agent. METHODS The target acetohydrazides (5a-m) were prepared via the Niementowski condensation of anthranilic acid (1a) or 5-substituted-2-aminobenzoic acid (1b-m) and formamide. The compound libraries were evaluated for their cytotoxicity, caspase-3 activation, cell cycle analysis, and apoptosis. In addition, computational chemistry is also performed. RESULTS A biological evaluation revealed that all thirteen compounds designed and synthesized showed strong cytotoxicity against three human cancer cell lines (SW620, colon cancer; PC-3, prostate cancer; NCI-H23, lung cancer) with eight compounds (5a, 5c-i, 5k), which were clearly more potent than both PAC-1 and oncrasin-1. In this series, four compounds including 5c, 5e, 5f, and 5h, were the most potent members with approximately 4- to 5-fold stronger than the reference compounds PAC-1 and oncrasin-1 in terms of IC50. In comparison to 5-FU, these compounds were even 18- to 29-fold more potent in terms of cytotoxicity in three human cell lines tested. In the caspase activation assay, the caspase activity was activated to 285% by compound 5e in comparison to PAC-1, the first procaspase activating compound, which was used as a control. Our docking simulation revealed that compound 5e was a potent allosteric inhibitor of procaspase-3 through chelation of inhibitory zinc ion. Physicochemical and ADMET calculations for 5e provided useful information of its suitable absorption profile and some toxicological effects that need further optimization to be developed as a promising anticancer agent. CONCLUSION Compound 5e has emerged as a potential hit for further design and development of caspases activators and anticancer agents.
Collapse
Affiliation(s)
- Do Thi Mai Dung
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Chungbuk, 28160, Republic of Korea
| | - Duong Tien Anh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Pham-The Hai
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Le Quang Bao
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - A Young Ji
- Department of Pharmacy, Chungbuk National University, Korea
| | - Jong Soon Kang
- Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, 28116, Republic of Korea
| | - Truong Thanh Tung
- PHENIKAA Institute for Advanced Study (PIAS), Phenikaa University, Hanoi 12116, Vietnam
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Chungbuk, 28160, Republic of Korea
| | - Nguyen-Hai Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| |
Collapse
|
13
|
Dung DTM, Park EJ, Anh DT, Hai PT, Huy LD, Jun HW, Kwon JH, Young Ji A, Kang JS, Tung TT, Dung PTP, Han SB, Nam NH. Design, synthesis, and evaluation of novel (E)-N'-(3-allyl-2-hydroxy)benzylidene-2-(4-oxoquinazolin-3(4H)-yl)acetohydrazides as antitumor agents. Arch Pharm (Weinheim) 2021; 355:e2100216. [PMID: 34674294 DOI: 10.1002/ardp.202100216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022]
Abstract
In our continuing search for novel small-molecule anticancer agents, we designed and synthesized a series of novel (E)-N'-(3-allyl-2-hydroxy)benzylidene-2-(4-oxoquinazolin-3(4H)-yl)acetohydrazides (5), focusing on the modification of substitution in the quinazolin-4(3H)-one moiety. The biological evaluation showed that all 13 designed and synthesized compounds displayed significant cytotoxicity against three human cancer cell lines (SW620, colon cancer; PC-3, prostate cancer; NCI-H23, lung cancer). The most potent compound 5l displayed cytotoxicity up to 213-fold more potent than 5-fluorouracil and 87-fold more potent than PAC-1, the first procaspase-activating compound. Structure-activity relationship analysis revealed that substitution of either electron-withdrawing or electron-releasing groups at positions 6 or 7 on the quinazolin-4(3H)-4-one moiety increased the cytotoxicity of the compounds, but substitution at position 6 seemed to be more favorable. In the caspase activation assay, compound 5l was found to activate the caspase activity by 291% in comparison to PAC-1, which was used as a control. Further docking simulation also revealed that this compound may be a potent allosteric inhibitor of procaspase-3 through chelation of the inhibitory zinc ion. Physicochemical and ADMET calculations for 5l provided useful information of its suitable absorption profile and some toxicological effects that need further optimization to be developed as a promising anticancer agent.
Collapse
Affiliation(s)
- Do T M Dung
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Eun J Park
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Duong T Anh
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Pham-The Hai
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Le D Huy
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Hye W Jun
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Joo-Hee Kwon
- Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, Republic of Korea
| | - A Young Ji
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jong S Kang
- Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, Republic of Korea
| | - Truong T Tung
- Faculty of Pharmacy, PHENIKAA University, Hanoi, Vietnam
- PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi, Vietnam
| | - Phan T P Dung
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Nguyen-Hai Nam
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| |
Collapse
|
14
|
El Mansouri AE, Oubella A, Mehdi A, AitItto MY, Zahouily M, Morjani H, Lazrek HB. Design, synthesis, biological evaluation and molecular docking of new 1,3,4-oxadiazole homonucleosides and their double-headed analogs as antitumor agents. Bioorg Chem 2021; 108:104558. [PMID: 33358270 DOI: 10.1016/j.bioorg.2020.104558] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/13/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
A novel series of homonucleosides and their double-headed analogs containing theophylline, 1,3,4-oxadiazole, and variant nucleobases was designed and synthesized. The new derivatives were fully characterized by HRMS, FT-IR, 1H NMR, and 13C NMR. The cytotoxic activities of all prepared compounds were screened in vitro against four cell lines, including fibrosarcoma (HT-1080), breast (MCF-7 and MDA-MB-231), and lung carcinoma (A-549). The double-headed analogue 18 showed marked growth inhibition against all the cell lines tested, specifically in HT-1080, with an IC50 values of 17.08 ± 0.97 µM. The possible mechanism of apoptosis was investigated using Annexin V staining, caspase-3/7 activity, and analysis cell cycle progression. The compound 18 induced apoptosis through caspase-3/7 activation and cell-cycle arrest in HT-1080 and A-549 cells. The molecular docking confirms that the compound 18 activated caspase-3 via the formation of hydrogen bonds and hydrophobic interactions.
Collapse
Affiliation(s)
- Az-Eddine El Mansouri
- Laboratoire de Materiaux, Catalyse & Valorisation des Ressources Naturelles, URAC 24, Faculte des Sciences et Techniques, Universite Hassan II, Casablanca, Morocco; Laboratory of Biomolecular and Medicinal Chemistry, Department of Chemistry, Faculty of Science Semlalia, BP 2390, Marrakech 40001, Morocco.
| | - Ali Oubella
- Laboratoire de Synthèse Organique et de Physico-Chimie Moléculaire, Département de Chimie, Faculte ́ des Sciences, Semlalia BP 2390, Marrakech 40001, Morocco
| | - Ahmad Mehdi
- Institut Charles Gerhardt Montpellier, UMR 5253, CNRS-UM-ENSCM, Université de Montpellier, Montpellier cedex 5, France
| | - Moulay Youssef AitItto
- Laboratoire de Synthèse Organique et de Physico-Chimie Moléculaire, Département de Chimie, Faculte ́ des Sciences, Semlalia BP 2390, Marrakech 40001, Morocco
| | - Mohamed Zahouily
- Laboratoire de Materiaux, Catalyse & Valorisation des Ressources Naturelles, URAC 24, Faculte des Sciences et Techniques, Universite Hassan II, Casablanca, Morocco; Moroccan Foundation for Advanced Science, Innovation and Research (MAScIR), VARENA Center, Rue Mohamed El Jazouli, Madinat Al Irfane, 10100 Rabat, Morocco.
| | - Hamid Morjani
- BioSpecT - EA7506 UFR de Pharmacie, Univ-Reims 51, rue Cognacq Jay, 51096 Reims cedex, France.
| | - Hassan B Lazrek
- Laboratory of Biomolecular and Medicinal Chemistry, Department of Chemistry, Faculty of Science Semlalia, BP 2390, Marrakech 40001, Morocco.
| |
Collapse
|
15
|
AkgÜl Ö, ErdoĞan MA, Bİrİm D, KayabaŞi Ç, GÜndÜz C, ArmaĞan G. Design, synthesis, cytotoxic activity, and apoptosis inducing effects of 4- and N-substituted benzoyltaurinamide derivatives. Turk J Chem 2021; 44:1674-1693. [PMID: 33488262 PMCID: PMC7763113 DOI: 10.3906/kim-2009-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/24/2020] [Indexed: 12/24/2022] Open
Abstract
In this study, a group of 4-substituted benzoyltaurinamide derivatives were designed, synthesized, and investigated for their anticancer activity against three cancer cell lines and one nontumorigenic cell line by MTT assay. Among the final compounds, methoxyphenyl derivatives 14, 15, 16 were found to be effective against all the tested cancerous cell lines with promising selectivity. The most active compounds were further evaluated to determine the molecular mechanism of their anticancer activity by using western blot assay and the Annexin V-FITC/PI test. Compound 14 (in SH-SY5Y and MDA-MB-231 cell lines) and 15 (in SH-SY5Y cell line) were found to induce intrinsic apoptotic pathway by upregulating BAX, caspase-3, and caspase-9, while downregulating Bcl-2 and Bcl-xL expression levels. According to mechanistic studies, compounds displayed their anticancer activity via three different mechanisms: a. caspase-dependent, b. caspase-independent, and c. caspase-dependent pathway that excluded caspase-9 activation. As a result, this study provides interesting data which can be used to design new taurine-based anticancer derivatives.
Collapse
Affiliation(s)
- Özlem AkgÜl
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, İzmir Turkey
| | - Mümin Alper ErdoĞan
- Department of Physiology, School of Medicine, İzmir Katip Çelebi University, İzmir Turkey
| | - Derviş Bİrİm
- Department of Biochemistry, Faculty of Pharmacy, Ege University, İzmir Turkey
| | - Çağla KayabaŞi
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir Turkey
| | - Cumhur GÜndÜz
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir Turkey
| | - Güliz ArmaĞan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, İzmir Turkey
| |
Collapse
|
16
|
Shao LH, Fan SL, Meng YF, Gan YY, Shao WB, Wang ZC, Chen DP, Ouyang GP. Design, synthesis, biological activities and 3D-QSAR studies of quinazolinone derivatives containing hydrazone structural units. NEW J CHEM 2021. [DOI: 10.1039/d0nj05450j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A series of novel quinazolinone derivatives containing hydrazone structural units were synthesized and their antitumour activities were evaluated.
Collapse
Affiliation(s)
- Li-Hui Shao
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
| | - Si-Li Fan
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
| | - Ying-Fen Meng
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
| | - Yi-Yuan Gan
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
- State Key Laboratory of Functions and Application of Medicinal Plants
| | - Wu-Bin Shao
- Center for R&D of Fine Chemicals of Guizhou University
- Guiyang
- China
| | - Zhen-Chao Wang
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
- State Key Laboratory of Functions and Application of Medicinal Plants
| | - Dan-Ping Chen
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
- State Key Laboratory of Functions and Application of Medicinal Plants
| | - Gui-Ping Ouyang
- College of Pharmacy
- Guizhou University
- Guiyang 550025
- China
- State Key Laboratory of Functions and Application of Medicinal Plants
| |
Collapse
|
17
|
Yang J, Huang D, Wang K, Wang J, Su Y, Deng Z, Yang T, Hu Y. Study on N-Alkylation Reactions of Trifluoromethylated Acylhydrazones. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202011001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Huan LC, Anh DT, Hai PT, Anh LD, Park EJ, Ji AY, Kang JS, Dung DTM, Oanh DTK, Tung TT, Hai DTT, Han SB, Nam NH. Design, synthesis, and evaluation of novel N'-substituted-1-(4-chlorobenzyl)-1 H-indol-3-carbohydrazides as antitumor agents. J Enzyme Inhib Med Chem 2020; 35:1854-1865. [PMID: 32981382 PMCID: PMC7534272 DOI: 10.1080/14756366.2020.1816997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In continuity of our search for novel anticancer agents acting as procaspase activators, we have designed and synthesised two series of (E)-N′-benzylidene-carbohydrazides (4a–m) and (Z)-N'-(2-oxoindolin-3-ylidene)carbohydrazides (5a–g) incorporating 1-(4-chlorobenzyl)-1H-indole core. Bioevaluation showed that the compounds, especially compounds in series 4a–m, exhibited potent cytotoxicity against three human cancer cell lines (SW620, colon cancer; PC-3, prostate cancer; NCI-H23, lung cancer). Within series 4a–m, compounds with 2-OH substituent (4g–i) exhibited very strong cytotoxicity in three human cancer cell lines assayed with IC50 values in the range of 0.56–0.83 µM. In particular, two compounds 4d and 4f bearing 4-Cl and 4-NO2 substituents, respectively, were the most potent in term of cytotoxicity with IC50 values of 0.011–0.001 µM. In caspase activation assay, compounds 4b and 4f were found to activate caspase activity by 314.3 and 270.7% relative to PAC-1. This investigation has demonstrated the potential of these simple acetohydrazides, especially compounds 4b, 4d, and 4f, as anticancer agents.
Collapse
Affiliation(s)
- Le Cong Huan
- Hanoi University of Pharmacy, Hanoi, Vietnam.,Thai Binh University of Medicine and Pharmacy, Thai Binh City, Vietnam
| | | | | | - Lai Duc Anh
- Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - A Young Ji
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Jong Soon Kang
- Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | | | | | - Truong Thanh Tung
- Faculty of Pharmacy, PHENIKAA University, Hanoi, Vietnam.,PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi, Vietnam
| | | | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | | |
Collapse
|
19
|
Li F, Han X, Chen Y, Wang S, Cheng Z, Hu G, Liu W, Zhu Q. In vitro metabolic characterization of orbitazine, a novel derivative of the PAC-1 anticancer agent. J Pharm Pharmacol 2020; 72:1199-1210. [PMID: 32583524 DOI: 10.1111/jphp.13296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/25/2020] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The in vitro evaluation of new drugs is an important step in the drug development pipeline. Orbitazine is a derivative of PAC-1 that has substituted the functional group homopiperazine ring with a piperazine ring. The purpose of this study was to assess the metabolic profile of orbitazine. METHODS Metabolism was characterized in vitro by incubating liver microsomes with metabolize orbitazine or the classical metabolic enzyme substrates. High performance liquid chromatography (HPLC) and LC-MS/MS were used to identify the parent drugs and metabolites of orbitazine or metabolic enzyme substrates. KEY FINDINGS There was no difference in metabolic stability or metabolites across different species. The metabolites included a debenzyl compound and several hydroxyl compounds, defined as M1(316), M2(440), M3(422), M4(422) and M5(422). We found that orbitazine was metabolized by CYP3A4, CYP2C9 and CYP2D6 in a human liver microsomes incubation system. Orbitazine had no significant inhibitory effect on CYP1A2, CYP2B6, CYP2C9, or CYP2C19 in human liver microsomes, but showed a dose-dependent inhibition of CYP2C8, CYP2D6 and CYP3A4; and there was no orbitazine-mediated induction of CYP1A2, CYP2B6, CYP3A4 or mRNA expression in hepatocytes. CONCLUSIONS This in vitro data on the metabolism of orbitazine may provide valuable information to support further clinical progression as a potential therapeutic molecule.
Collapse
Affiliation(s)
- Fang Li
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Xuhua Han
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Yanfen Chen
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Shanshan Wang
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Zeneng Cheng
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Gaoyun Hu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Wenjie Liu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Huan LC, Anh DT, Truong BX, Duc PH, Hai PT, Duc-Anh L, Huong LTT, Park EJ, Lee HJ, Kang JS, Tran PT, Thanh Hai DT, Kim Oanh DT, Han SB, Nam NH. New Acetohydrazides Incorporating 2-Oxoindoline and 4-Oxoquinazoline: Synthesis and Evaluation of Cytotoxicity and Caspase Activation Activity. Chem Biodivers 2020; 17:e1900670. [PMID: 31943757 DOI: 10.1002/cbdv.201900670] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
In our search for new small molecules activating procaspase-3, we have designed and synthesized a series of new acetohydrazides incorporating both 2-oxoindoline and 4-oxoquinazoline scaffolds. Biological evaluation showed that a number of these acetohydrazides were comparably or even more cytotoxic against three human cancer cell lines (SW620, colon cancer; PC-3, prostate cancer; NCI-H23, lung cancer) in comparison to PAC-1, a first procaspase-3 activating compound, which was used as a positive control. One of those new compounds, 2-(6-chloro-4-oxoquinazolin-3(4H)-yl)-N'-[(3Z)-5-methyl-2-oxo-1,2-dihydro-3H-indol-3-ylidene]acetohydrazide activated the caspase-3 activity in U937 human lymphoma cells by 5-fold higher than the untreated control. Three of the new compounds significantly induced necrosis and apoptosis in U937 cells.
Collapse
Affiliation(s)
- Le Cong Huan
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Duong Tien Anh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Bui Xuan Truong
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Phan Huy Duc
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Pham-The Hai
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Lai Duc-Anh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Le-Thi-Thu Huong
- School of Medicine and Pharmacy, Hanoi National University, 144 Xuan Thuy, Hanoi, 10000, Vietnam
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Hye Jin Lee
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jong Soon Kang
- Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, 28116, Republic of Korea
| | - Phuong-Thao Tran
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Dinh Thi Thanh Hai
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Dao Thi Kim Oanh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Nguyen-Hai Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 10000, Vietnam
| |
Collapse
|
21
|
Boudreau MW, Peh J, Hergenrother PJ. Procaspase-3 Overexpression in Cancer: A Paradoxical Observation with Therapeutic Potential. ACS Chem Biol 2019; 14:2335-2348. [PMID: 31260254 PMCID: PMC6858495 DOI: 10.1021/acschembio.9b00338] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many anticancer strategies rely on the promotion of apoptosis in cancer cells as a means to shrink tumors. Crucial for apoptotic function are executioner caspases, most notably caspase-3, that proteolyze a variety of proteins, inducing cell death. Paradoxically, overexpression of procaspase-3 (PC-3), the low-activity zymogen precursor to caspase-3, has been reported in a variety of cancer types. Until recently, this counterintuitive overexpression of a pro-apoptotic protein in cancer has been puzzling. Recent studies suggest subapoptotic caspase-3 activity may promote oncogenic transformation, a possible explanation for the enigmatic overexpression of PC-3. Herein, the overexpression of PC-3 in cancer and its mechanistic basis is reviewed; collectively, the data suggest the potential for exploitation of PC-3 overexpression with PC-3 activators as a targeted anticancer strategy.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| |
Collapse
|
22
|
Huan LC, Tran PT, Phuong CV, Duc PH, Anh DT, Hai PT, Huong LTT, Thuan NT, Lee HJ, Park EJ, Kang JS, Linh NP, Hieu TT, Oanh DTK, Han SB, Nam NH. Novel 3,4-dihydro-4-oxoquinazoline-based acetohydrazides: Design, synthesis and evaluation of antitumor cytotoxicity and caspase activation activity. Bioorg Chem 2019; 92:103202. [DOI: 10.1016/j.bioorg.2019.103202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 02/02/2023]
|
23
|
Lin A, Giuliano CJ, Palladino A, John KM, Abramowicz C, Yuan ML, Sausville EL, Lukow DA, Liu L, Chait AR, Galluzzo ZC, Tucker C, Sheltzer JM. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci Transl Med 2019; 11:eaaw8412. [PMID: 31511426 PMCID: PMC7717492 DOI: 10.1126/scitranslmed.aaw8412] [Citation(s) in RCA: 429] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/19/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
Ninety-seven percent of drug-indication pairs that are tested in clinical trials in oncology never advance to receive U.S. Food and Drug Administration approval. While lack of efficacy and dose-limiting toxicities are the most common causes of trial failure, the reason(s) why so many new drugs encounter these problems is not well understood. Using CRISPR-Cas9 mutagenesis, we investigated a set of cancer drugs and drug targets in various stages of clinical testing. We show that-contrary to previous reports obtained predominantly with RNA interference and small-molecule inhibitors-the proteins ostensibly targeted by these drugs are nonessential for cancer cell proliferation. Moreover, the efficacy of each drug that we tested was unaffected by the loss of its putative target, indicating that these compounds kill cells via off-target effects. By applying a genetic target-deconvolution strategy, we found that the mischaracterized anticancer agent OTS964 is actually a potent inhibitor of the cyclin-dependent kinase CDK11 and that multiple cancer types are addicted to CDK11 expression. We suggest that stringent genetic validation of the mechanism of action of cancer drugs in the preclinical setting may decrease the number of therapies tested in human patients that fail to provide any clinical benefit.
Collapse
Affiliation(s)
- Ann Lin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Christopher J Giuliano
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Ann Palladino
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kristen M John
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Hofstra University, Hempstead, NY 11549, USA
| | - Connor Abramowicz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- New York Institute of Technology, Glen Head, NY 11545, USA
| | - Monet Lou Yuan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Syosset High School, Syosset, NY 11791, USA
| | - Erin L Sausville
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Devon A Lukow
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Luwei Liu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | - Clara Tucker
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Jason M Sheltzer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
24
|
Yurttaş L, Öztürk Ö, Cantürk Z. New Procaspase Activating Compound (PAC-1) Like Molecules as Potent Antitumoral Agents Against Lung Cancer. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666180926113040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
In this study, novel ortho-hydroxy N-acyl hydrazone moiety including
compounds (3a-l) were designed, based on procaspase activating compound (PAC-1) which is a
small molecule known with antitumor activity. The antitumor activity was evaluated on A549 (human
lung cancer cell line) and CCD 19Lu (human lung normal cell line).
Methods:
Twelve N'-arylidene-2-[4-(methylsulfonyl)piperazin-1-yl]acetohydrazide derivatives
(3a-l) were synthesized starting from ethyl 1-piperazinylacetate. All compounds were tested using
MTT method and Xcelligence-Real time cell analysis system (RTCA DP) to determine their antitumor
activity.
Results:
Some physicochemical properties of four active compounds were also predicted using
MolSoft, PreADMET and PROTOX software. Four of them, 3h, 3j, 3k and 3l bearing 3-hydroxy,
4-dimethylamino, 2,6-dichloro and 3,4-dichloro substituents in order exhibited selective cytotoxicity.
Conclusion:
Eligible values were obtained in the specified ranges as to be an oral/intravenous drug
considering the physicochemical calculations.
Collapse
Affiliation(s)
- Leyla Yurttaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
| | - Ömer Öztürk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
| | - Zerrin Cantürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
| |
Collapse
|
25
|
Huan LC, Phuong CV, Truc LC, Thanh VN, Pham-The H, Huong LTT, Thuan NT, Park EJ, Ji AY, Kang JS, Han SB, Tran PT, Nam NH. (E)-N'-Arylidene-2-(4-oxoquinazolin-4(3H)-yl) acetohydrazides: Synthesis and evaluation of antitumor cytotoxicity and caspase activation activity. J Enzyme Inhib Med Chem 2019; 34:465-478. [PMID: 30734614 PMCID: PMC6338265 DOI: 10.1080/14756366.2018.1555536] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In our search for novel small molecules activating procaspase-3, we have designed and synthesised a series of novel acetohydrazides incorporating quinazolin-4(3H)-ones (5, 6, 7). Biological evaluation revealed eight compounds with significant cytotoxicity against three human cancer cell lines (SW620, colon cancer; PC-3, prostate cancer; NCI-H23, lung cancer). The most potent compound 5t displayed cytotoxicity up to 5-fold more potent than 5-FU. Analysis of structure-activity relationships showed that the introduction of different substituents at C-6 position on the quinazolin-4(3H)-4-one moiety, such as 6-chloro or 6-methoxy potentially increased the cytotoxicity of the compounds. In term of caspase activation activity, several compounds were found to exhibit potent effects, (e.g. compounds 7 b, 5n, and 5l). Especially, compound 7 b activated caspases activity by almost 200% in comparison to that of PAC-1. Further docking simulation also revealed that this compound potentially is a potent allosteric inhibitor of procaspase-3.
Collapse
Affiliation(s)
- Le Cong Huan
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Cao Viet Phuong
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Le Cong Truc
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Vo Nguyen Thanh
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Hai Pham-The
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Le-Thi-Thu Huong
- b School of Medicine and Pharmacy , Vietnam National University , Hanoi , Vietnam
| | - Nguyen Thi Thuan
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Eun Jae Park
- c College of Pharmacy , Chungbuk National University , Cheongju , Republic of Korea
| | - A Young Ji
- c College of Pharmacy , Chungbuk National University , Cheongju , Republic of Korea
| | - Jong Soon Kang
- d Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology , Cheongju , Republic of Korea
| | - Sang-Bae Han
- c College of Pharmacy , Chungbuk National University , Cheongju , Republic of Korea
| | - Phuong-Thao Tran
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| | - Nguyen-Hai Nam
- a Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| |
Collapse
|
26
|
Huan LC, Truc LC, Phuong CV, Hai PT, Huong LTT, Linh NTP, Thuan NT, Park EJ, Choi YJ, Kang JS, Han SB, Nam NH, Tran PT. N'-[(E)-Arylidene]-2-(2,3-dihydro-3-oxo-4H-1,4-benzoxazin-4-yl)-acetohydrazides: Synthesis and Evaluation of Caspase Activation Activity and Cytotoxicity. Chem Biodivers 2018; 15:e1800322. [PMID: 30054973 DOI: 10.1002/cbdv.201800322] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/25/2018] [Indexed: 11/08/2022]
Abstract
In our search for novel small cytotoxic molecules potentially activating procaspase-3, we have designed and synthesized a series of novel N'-[(E)-arylidene]-2-(2,3-dihydro-3-oxo-4H-1,4-benzoxazin-4-yl)acetohydrazides (5, 6). Biological evaluation revealed that seven compounds, including 5h, 5j, 5k, 5l, 5n, 6a, and 6b, exhibited moderate to strong cytotoxicity against three human cancer cell lines (SW620, colon cancer; PC-3, prostate cancer; NCI-H23, lung cancer). Among these compounds, two most cytotoxic compounds (5h and 5j) displayed from 3- up to 10-fold higher potency than PAC-1 and 5-FU in three cancer cell lines tested. Three compounds 5j, 5k, and 5n were also found to display better caspases activation activity in comparison to PAC-1. Especially, compound 5k activated the level of caspases activity by 200% higher than that of PAC-1. From this study, three compounds 5j, 5k, and 5n could be considered as potential leads for further design and development of caspase activators and anticancer agents.
Collapse
Affiliation(s)
- Le Cong Huan
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Le Cong Truc
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Cao Viet Phuong
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Pham-The Hai
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Le-Thi-Thu Huong
- School of Medicine and Pharmacy, Vietnam National University, 144 Xuan Thuy, Hanoi, Vietnam
| | | | - Nguyen Thi Thuan
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Yeo Jin Choi
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jong Soon Kang
- Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyung-1, Heungdeok, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Nguyen-Hai Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Phuong-Thao Tran
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| |
Collapse
|
27
|
Said B, Montenegro I, Valenzuela M, Olguín Y, Caro N, Werner E, Godoy P, Villena J, Madrid A. Synthesis and Antiproliferative Activity of New Cyclodiprenyl Phenols against Select Cancer Cell Lines. Molecules 2018; 23:molecules23092323. [PMID: 30213053 PMCID: PMC6225466 DOI: 10.3390/molecules23092323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/31/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023] Open
Abstract
Six new cyclodiprenyl phenols were synthesized by direct coupling of perillyl alcohol and the appropriate phenol. Their structures were established by IR, HRMS and mainly NMR. Three human cancer cell lines-breast (MCF-7), prostate (PC-3) and colon (HT-29)-were used in antiproliferative assays, with daunorubicin and dunnione as positive controls. Results described in the article suggest that dihydroxylated compounds 2⁻4 and monohydroxylated compound 5 display selectivity against cancer cell lines, cytotoxicity, apoptosis induction, and mitochondrial membrane impairment capacity. Compound 2 was identified as the most effective of the series by displaying against all cancer cell lines a cytotoxicity close to dunnione antineoplastic agent, suggesting that the cyclodiprenyl phenols from perillyl alcohol deserve more extensive investigation of their potential medicinal applications.
Collapse
Affiliation(s)
- Bastián Said
- Departamento de Química, Universidad Técnica Federico Santa María, Av. Santa María 6400, Vitacura 7630000, Santiago, Chile.
| | - Iván Montenegro
- Escuela de Obstetricia y Puericultura, Facultad de medicina, Campus de la Salud, Universidad de Valparaíso, Angamos 655, Reñaca, Viña del Mar 2520000, Chile.
| | - Manuel Valenzuela
- Laboratorio de Microbiología Celular, Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8320000, Chile.
| | - Yusser Olguín
- Center for Integrative Medicine and Innovative Science (CIMIS), Facultad de Medicina, Universidad Andrés Bello, Santiago 8320000, Chile.
| | - Nelson Caro
- Centro de Investigación Australbiotech, Universidad Santo Tomás, Avda. Ejército 146, Santiago 8320000, Chile.
| | - Enrique Werner
- Departamento De Ciencias Básicas, Campus Fernando May Universidad del Biobío, Avda. Andrés Bello s/n casilla 447, Chillán 3780000, Chile.
| | - Patricio Godoy
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Los Laureles s/n, Isla Teja, Valdivia 5090000, Chile.
| | - Joan Villena
- Centro de Investigaciones Biomedicas (CIB), Facultad de Medicina, Campus de la Salud, Universidad de Valparaíso, Angamos 655, Reñaca, Viña del Mar 2520000, Chile.
| | - Alejandro Madrid
- Laboratorio de Productos Naturales y Síntesis Orgánica, Departamento de Química, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Avda. Leopoldo Carvallo 270, Playa Ancha, Valparaíso 2340000, Chile.
| |
Collapse
|
28
|
Peh J, Boudreau MW, Smith HM, Hergenrother PJ. Overcoming Resistance to Targeted Anticancer Therapies through Small-Molecule-Mediated MEK Degradation. Cell Chem Biol 2018; 25:996-1005.e4. [PMID: 29909991 PMCID: PMC6097934 DOI: 10.1016/j.chembiol.2018.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/31/2018] [Accepted: 05/04/2018] [Indexed: 02/09/2023]
Abstract
The discovery of mutant or fusion kinases that drive oncogenesis, and the subsequent approval of specific inhibitors for these enzymes, has been instrumental in the management of some cancers. However, acquired resistance remains a significant problem in the clinic, limiting the long-term effectiveness of most of these drugs. Here we demonstrate a general strategy to overcome this resistance through drug-induced MEK cleavage (via direct procaspase-3 activation) combined with targeted kinase inhibition. This combination effect is shown to be general across diverse tumor histologies (melanoma, lung cancer, and leukemia) and driver mutations (mutant BRAF or EGFR, fusion kinases EML4-ALK and BCR-ABL). Caspase-3-mediated degradation of MEK kinases results in sustained pathway inhibition and substantially delayed or eliminated resistance in cancer cells in a manner far superior to combinations with MEK inhibitors. These data suggest the generality of drug-mediated MEK kinase cleavage as a therapeutic strategy to prevent resistance to targeted anticancer therapies.
Collapse
Affiliation(s)
- Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 261 Roger Adams Lab Box 36-5, 600 S. Mathews Avenue, Urbana, IL 61801, USA
| | - Matthew W Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 261 Roger Adams Lab Box 36-5, 600 S. Mathews Avenue, Urbana, IL 61801, USA
| | - Hannah M Smith
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 261 Roger Adams Lab Box 36-5, 600 S. Mathews Avenue, Urbana, IL 61801, USA
| | - Paul J Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 261 Roger Adams Lab Box 36-5, 600 S. Mathews Avenue, Urbana, IL 61801, USA.
| |
Collapse
|
29
|
Structural design, synthesis and substituent effect of hydrazone-N-acylhydrazones reveal potent immunomodulatory agents. Bioorg Med Chem 2018. [DOI: 10.1016/j.bmc.2018.02.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Fulda S. Therapeutic opportunities based on caspase modulation. Semin Cell Dev Biol 2017; 82:150-157. [PMID: 29247787 DOI: 10.1016/j.semcdb.2017.12.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
Abstract
Caspases are a family of proteolytic enzymes that play a critical role in the regulation of programmed cell death via apoptosis. Activation of caspases is frequently impaired in human cancers, contributing to cancer formation, progression and therapy resistance. A better understanding of the molecular mechanisms regulating caspase activation in cancer cells is therefore highly important. Thus, targeted modulation of caspase activation and apoptosis represents a promising approach for the development of new therapeutic options to elucidate cancer cell death.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
31
|
Joshi AD, Botham RC, Schlein LJ, Roth HS, Mangraviti A, Borodovsky A, Tyler B, Joslyn S, Looper JS, Podell M, Fan TM, Hergenrother PJ, Riggins GJ. Synergistic and targeted therapy with a procaspase-3 activator and temozolomide extends survival in glioma rodent models and is feasible for the treatment of canine malignant glioma patients. Oncotarget 2017; 8:80124-80138. [PMID: 29113289 PMCID: PMC5655184 DOI: 10.18632/oncotarget.19085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/09/2017] [Indexed: 12/17/2022] Open
Abstract
Purpose Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. Experimental Design Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. Results Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. Conclusions Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Avadhut D Joshi
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel C Botham
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Lisa J Schlein
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Howard S Roth
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Antonella Mangraviti
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alexandra Borodovsky
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Jayme S Looper
- Department of Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Michael Podell
- Department of Neurology, MedVet Chicago, Chicago, IL, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Gregory J Riggins
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Weekley CM, He C. Developing drugs targeting transition metal homeostasis. Curr Opin Chem Biol 2016; 37:26-32. [PMID: 28040658 DOI: 10.1016/j.cbpa.2016.12.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/23/2016] [Accepted: 12/08/2016] [Indexed: 01/06/2023]
Abstract
Metal dyshomeostasis is involved in the pathogenesis and progression of diseases including cancer and neurodegenerative diseases. Metal chelators and ionophores are well known modulators of transition metal homeostasis, and a number of these molecules are in clinical trials. Metal-binding compounds are not the only drugs capable of targeting transition metal homeostasis. This review presents recent highlights in the development of chelators and ionophores for the treatment of cancer and neurodegenerative disease. Moreover, we discuss the development of small molecules that alter copper and iron homeostasis by inhibiting metal transport proteins. Finally, we consider the emergence of metal regulatory factor 1 as a drug target in diseases where it mediates zinc-induced signalling cascades leading to pathogenesis.
Collapse
Affiliation(s)
- Claire M Weekley
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
33
|
Botham R, Roth HS, Book AP, Roady PJ, Fan TM, Hergenrother PJ. Small-Molecule Procaspase-3 Activation Sensitizes Cancer to Treatment with Diverse Chemotherapeutics. ACS CENTRAL SCIENCE 2016; 2:545-59. [PMID: 27610416 PMCID: PMC4999974 DOI: 10.1021/acscentsci.6b00165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Indexed: 05/08/2023]
Abstract
Conventional chemotherapeutics remain essential treatments for most cancers, but their combination with other anticancer drugs (including targeted therapeutics) is often complicated by unpredictable synergies and multiplicative toxicities. As cytotoxic anticancer chemotherapeutics generally function through induction of apoptosis, we hypothesized that a molecularly targeted small molecule capable of facilitating a central and defining step in the apoptotic cascade, the activation of procaspase-3 to caspase-3, would broadly and predictably enhance activity of cytotoxic drugs. Here we show that procaspase-activating compound 1 (PAC-1) enhances cancer cell death induced by 15 different FDA-approved chemotherapeutics, across many cancer types and chemotherapeutic targets. In particular, the promising combination of PAC-1 and doxorubicin induces a synergistic reduction in tumor burden and enhances survival in murine tumor models of osteosarcoma and lymphoma. This PAC-1/doxorubicin combination was evaluated in 10 pet dogs with naturally occurring metastatic osteosarcoma or lymphoma, eliciting a biologic response in 3 of 6 osteosarcoma patients and 4 of 4 lymphoma patients. Importantly, in both mice and dogs, coadministration of PAC-1 with doxorubicin resulted in no additional toxicity. On the basis of the mode of action of PAC-1 and the high expression of procaspase-3 in many cancers, these results suggest the combination of PAC-1 with cytotoxic anticancer drugs as a potent and general strategy to enhance therapeutic response.
Collapse
Affiliation(s)
- Rachel
C. Botham
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Howard S. Roth
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Alison P. Book
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Patrick J. Roady
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
- E-mail:
| |
Collapse
|
34
|
Peh J, Fan TM, Wycislo KL, Roth HS, Hergenrother PJ. The Combination of Vemurafenib and Procaspase-3 Activation Is Synergistic in Mutant BRAF Melanomas. Mol Cancer Ther 2016; 15:1859-69. [PMID: 27297867 DOI: 10.1158/1535-7163.mct-16-0025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/31/2016] [Indexed: 12/14/2022]
Abstract
The development of vemurafenib resistance limits the long-term efficacy of this drug for treatment of metastatic melanomas with the (V600E)BRAF mutation. Inhibition of downstream MAPK signaling with vemurafenib induces apoptotic cell death mediated by caspase-3, suggesting that addition of a procaspase-3 activator could enhance anticancer effects. Here, we show that the combination of PAC-1, a procaspase-activating compound, and vemurafenib is highly synergistic in enhancing caspase-3 activity and apoptotic cell death in melanoma cell lines harboring the (V600E)BRAF mutation. In vivo, the combination displays a favorable safety profile in mice and exerts significant antitumor effects. We further demonstrate that addition of PAC-1 to the clinically useful combination of vemurafenib and a MEK inhibitor, trametinib, starkly enhances the caspase-3 activity and proapoptotic effect of the combination. Moreover, addition of low concentration PAC-1 also delays the regrowth of cells following treatment with vemurafenib. Finally, PAC-1 remains potent against vemurafenib-resistant A375VR cells in cell culture and synergizes with vemurafenib to exert antitumor effects on A375VR cell growth in vivo Collectively, our data suggest that inhibition of MAPK signaling combined with concurrent procaspase-3 activation is an effective strategy to enhance the antitumor activity of vemurafenib and mitigate the development of resistance. Mol Cancer Ther; 15(8); 1859-69. ©2016 AACR.
Collapse
Affiliation(s)
- Jessie Peh
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois. Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Timothy M Fan
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois. Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Kathryn L Wycislo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Howard S Roth
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois. Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois. Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|