1
|
Braga CB, Perli G, Fonseca R, Grigolo TA, Ionta M, Ornelas C, Pilli RA. Enhanced Synergistic Efficacy Against Breast Cancer Cells Promoted by Co-Encapsulation of Piplartine and Paclitaxel in Acetalated Dextran Nanoparticles. Mol Pharm 2024; 21:5577-5597. [PMID: 39365693 DOI: 10.1021/acs.molpharmaceut.4c00548] [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] [Indexed: 10/06/2024]
Abstract
Malignant breast tumors constitute the most frequent cancer diagnosis among women. Notwithstanding the progress in treatments, this condition persists as a major public health issue. Paclitaxel (PTX) is a first-line classical chemotherapeutic drug used as a single active pharmaceutical ingredient (API) or in combination therapy for breast cancer (BC) treatment. Adverse effects, poor water solubility, and inevitable susceptibility to drug resistance seriously limit its therapeutic efficacy in the clinic. Piplartine (PPT), an alkaloid extracted from Piper longum L., has been shown to inhibit cancer cell proliferation in several cell lines due to its pro-oxidant activity. However, PPT has low water solubility and bioavailability in vivo, and new strategies should be developed to optimize its use as a chemotherapeutic agent. In this context, the present study aimed to synthesize a series of acetalated dextran nanoparticles (Ac-Dex NPs) encapsulating PPT and PTX to overcome the limitations of PPT and PTX, maximizing their therapeutic efficacy and achieving prolonged and targeted codelivery of these anticancer compounds into BC cells. Biodegradable, pH-responsive, and biocompatible Ac-Dex NPs with diameters of 100-200 nm and spherical morphologies were formulated using a single emulsion method. Selected Ac-Dex NPs containing only PPT or PTX as well as those coloaded with PPT and PTX achieved excellent drug-loading capabilities (PPT, ca. 11-33%; PTX, ca. 2-14%) and high encapsulation efficiencies (PPT, ∼57-98%; PTX, ∼80-97%). Under physiological conditions (pH 7.4), these NPs exhibited excellent colloidal stability and were capable of protecting drug release, while under acidic conditions (pH 5.5) they showed structural collapse, releasing the therapeutics in an extended manner. Cytotoxicity results demonstrated that the encapsulation in Ac-Dex NPs had a positive effect on the activities of both PPT and PTX against the MCF-7 human breast cancer cell line after 48 h of treatment, as well as toward MDA-MB-231 triple-negative BC cells. PPT/PTX@Ac-Dex NPs were significantly more cytotoxic (IC50/PPT = 0.25-1.77 μM and IC50/PTX = 0.07-0.75 μM) and selective (SI = 2.9-6.7) against MCF-7 cells than all the control therapeutic agents: free PPT (IC50 = 4.57 μM; SI = 1.2), free PTX (IC50 = 0.97 μM; SI = 1.0), the single-drug-loaded Ac-Dex NPs, and the physical mixture of both free drugs. All combinations of PPT and PTX resulted in pronounced synergistic antiproliferative effects in MCF-7 cells, with an optimal molar ratio of PPT to PTX of 2.3:1. PPT/PTX-2@Ac-Dex NPs notably promoted apoptosis, cell cycle arrest at the G2/M, accumulation of intracellular reactive oxygen species (ROS), and combined effects from both PPT and PTX on the microtubule network of MCF-7 cells. Overall, the combination of PTX and PPT in pH-responsive Ac-Dex NPs may offer great potential to improve the therapeutic efficacy, overcome the limitations, and provide effective simultaneous delivery of these therapeutics for BC treatment.
Collapse
Affiliation(s)
- Carolyne Brustolin Braga
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, P.O. Box 6154, 13083-970, Campinas, São Paulo Brazil
| | - Gabriel Perli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, P.O. Box 6154, 13083-970, Campinas, São Paulo Brazil
- POLYMAT, University of the Basque Country UPV/EHU, Joxe Mari Korta Center, 20018 Donostia-San Sebastián Spain
| | - Rafael Fonseca
- Institute of Biomedical Sciences, Federal University of Alfenas, UNIFAL-MG, 37130-001 Alfenas, Minas Gerais, Brazil
| | - Thiago Augusto Grigolo
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, P.O. Box 6154, 13083-970, Campinas, São Paulo Brazil
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Alfenas, UNIFAL-MG, 37130-001 Alfenas, Minas Gerais, Brazil
| | - Catia Ornelas
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, P.O. Box 6154, 13083-970, Campinas, São Paulo Brazil
- R&D Department, ChemistryX, R&D and Consulting Company, 9000 Funchal, Portugal
- R&D Department, Dendriwave, Research & Development Start-Up Company, 9000 Funchal, Portugal
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, P.O. Box 6154, 13083-970, Campinas, São Paulo Brazil
| |
Collapse
|
2
|
Jiang Y, Wang C, Zu C, Rong X, Yu Q, Jiang J. Synergistic Potential of Nanomedicine in Prostate Cancer Immunotherapy: Breakthroughs and Prospects. Int J Nanomedicine 2024; 19:9459-9486. [PMID: 39371481 PMCID: PMC11456300 DOI: 10.2147/ijn.s466396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Given the global prevalence of prostate cancer in men, it is crucial to explore more effective treatment strategies. Recently, immunotherapy has emerged as a promising cancer treatment due to its unique mechanism of action and potential long-term effectiveness. However, its limited efficacy in prostate cancer has prompted renewed interest in developing strategies to improve immunotherapy outcomes. Nanomedicine offers a novel perspective on cancer treatment with its unique size effects and surface properties. By employing targeted delivery, controlled release, and enhanced immunogenicity, nanoparticles can be synergized with nanomedicine platforms to amplify the effectiveness of immunotherapy in treating prostate cancer. Simultaneously, nanotechnology can address the limitations of immunotherapy and the challenges of immune escape and tumor microenvironment regulation. Additionally, the synergistic effects of combining nanomedicine with other therapies offer promising clinical outcomes. Innovative applications of nanomedicine include smart nanocarriers, stimulus-responsive systems, and precision medicine approaches to overcome translational obstacles in prostate cancer immunotherapy. This review highlights the transformative potential of nanomedicine in enhancing prostate cancer immunotherapy and emphasizes the need for interdisciplinary collaboration to drive research and clinical applications forward.
Collapse
Affiliation(s)
- Yueyao Jiang
- Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Chuancheng Zu
- China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Qian Yu
- Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| |
Collapse
|
3
|
Sankar DR, Neetha M, Anilkumar G. Gold-Catalyzed Lactone Synthesis: Advancements and Insights. CHEM REC 2024; 24:e202400071. [PMID: 39051735 DOI: 10.1002/tcr.202400071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/03/2024] [Indexed: 07/27/2024]
Abstract
Lactones represent a class of fundamental structural motifs ubiquitous in nature, holding significance across diverse scientific domains such as pharmaceuticals, natural products, drug discovery, and industry. Despite their simplicity, the synthesis of lactones has garnered considerable interest due to their pivotal roles. Gold, traditionally regarded as a noble metal, has emerged as an efficient catalyst, challenging conventional perceptions. The utilization of gold in lactone synthesis has captivated researchers, leading to the development of numerous effective methodologies. Motivated by this, we present a comprehensive compilation of reports on the gold-catalyzed synthesis of lactones, encompassing literature till date.
Collapse
Affiliation(s)
- D Ravi Sankar
- Department of Chemistry, Sree Narayana College, Sreenivasapuram, Varkala, Kerala, INDIA, 695145
| | - Mohan Neetha
- School of Chemical Sciences, Mahatma Gandhi University, Priyadarsini Hills P. O., Kottayam, Kerala, INDIA, 686560
| | - Gopinathan Anilkumar
- School of Chemical Sciences, Mahatma Gandhi University, Priyadarsini Hills P. O., Kottayam, Kerala, INDIA, 686560
- Institute for Integrated programs and Research in Basic Sciences (IIRBS), Mahatma Gandhi University, Priyadarsini Hills P. O., Kottayam, Kerala, INDIA, 686560
| |
Collapse
|
4
|
Pradhan D, Biswasroy P, Ramchandani M, Pradhan DK, Bhola RK, Goyal A, Ghosh G, Rath G. Development, characterization, and evaluation of withaferin-A and artesunate-loaded pH-responsive acetal-dextran polymeric nanoparticles for the management of malaria. Int J Biol Macromol 2024; 273:133220. [PMID: 38897506 DOI: 10.1016/j.ijbiomac.2024.133220] [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: 07/31/2023] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Artemisinin and its derivatives have been commonly used to treat malaria. However, the emergence of resistance against artemisinin derivatives has posed a critical challenge in malaria management. In the present study, we have proposed a combinatorial approach, utilizing pH-responsive acetal-dextran nanoparticles (Ac-Dex NPs) as carriers for the delivery of withaferin-A (WS-3) and artesunate (Art) to improve treatment efficacy of malaria. The optimized WS-3 and Art Ac-Dex NPs demonstrated enhanced pH-responsive release profiles under parasitophorous mimetic conditions (pH 5.5). Computational molecular modeling reveals that Ac-Dex's polymeric backbone strongly interacts with merozoite surface protein-1 (MSP-1), preventing erythrocyte invasion. In-vitro antimalarial activity of drug-loaded Ac-Dex NPs reveals a 1-1.5-fold reduction in IC50 values compared to pure drug against the 3D7 strain of Plasmodium falciparum. Treatment with WS-3 Ac-Dex NPs (100 mg/kg) and Art Ac-Dex NPs (30 mg/kg) to Plasmodium berghei-infected mice resulted in 78.11 % and 100 % inhibition of parasitemia. Notably, the combination therapy comprised of Art and WS-3 Ac-Dex NPs achieved complete inhibition of parasitemia even at a half dose of Art, indicating the synergistic potential of the combinations. However, further investigations are necessary to confirm the safety and effectiveness of WS-3 and Art Ac-Dex NPs for their successful clinical implications.
Collapse
Affiliation(s)
- Deepak Pradhan
- Department of Herbal Nanotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Prativa Biswasroy
- Department of Herbal Nanotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Manish Ramchandani
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, Rajasthan, India
| | - Dilip Kumar Pradhan
- Department of Medicine, Pandit Raghunath Murmu Medical College and Hospital, Baripada, Odisha, India
| | - Rajesh Kumar Bhola
- Department of Hematology, Institute of Medical Sciences and Sum Hospital, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Amit Goyal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, Rajasthan, India
| | - Goutam Ghosh
- Department of Herbal Nanotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India.
| | - Goutam Rath
- Department of Herbal Nanotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India.
| |
Collapse
|
5
|
Haffez H, Elsayed NA, Ahmed MF, Fatahala SS, Khaleel EF, Badi RM, Elkaeed EB, El Hassab MA, Hammad SF, Eldehna WM, Masurier N, El-Haggar R. Novel N-Arylmethyl-aniline/chalcone hybrids as potential VEGFR inhibitors: synthesis, biological evaluations, and molecular dynamic simulations. J Enzyme Inhib Med Chem 2023; 38:2278022. [PMID: 37982203 PMCID: PMC11003488 DOI: 10.1080/14756366.2023.2278022] [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: 08/25/2023] [Accepted: 10/25/2023] [Indexed: 11/21/2023] Open
Abstract
Significant advancements have been made in the domain of targeted anticancer therapy for the management of malignancies in recent times. VEGFR-2 is characterised by its pivotal involvement in angiogenesis and subsequent mechanisms that promote tumour cells survival. Herein, novel N-arylmethyl-aniline/chalcone hybrids 5a-5n were designed and synthesised as potential anticancer and VEGFR-2 inhibitors. The anticancer activity was evaluated at the NCI-USA, resulting in the identification of 10 remarkably potent molecules 5a-5j that were further subjected to the five-dose assays. Thereafter, they were explored for their VEGFR-2 inhibitory activity where 5e and 5h emerged as the most potent inhibitors. 5e and 5h induced apoptosis with cell cycle arrest at the SubG0-G1 phase within HCT-116 cells. Moreover, their impact on some key apoptotic genes was assessed, suggesting caspase-dependent apoptosis. Furthermore, molecular docking and molecular dynamics simulations were conducted to explore the binding modes and stability of the protein-ligand complexes.
Collapse
Affiliation(s)
- Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo, Egypt
| | - Nosaiba A. Elsayed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
| | - Marwa F. Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
| | - Samar S. Fatahala
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Eman F. Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Rehab Mustafa Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mahmoud A. El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt
| | - Sherif F. Hammad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
- Medicinal Chemistry Department, PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, Egypt Alexandria
| | - Wagdy M. Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Nicolas Masurier
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Radwan El-Haggar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
| |
Collapse
|
6
|
Abubakar A, Abdulmalek E, Norhamidah Wan Ibrahim W, Cordova KE, Abdul Rahman MB. ZIF-90 nanoparticles modified with a homing peptide for targeted delivery of cisplatin. Front Chem 2022; 10:1076350. [PMID: 36545218 PMCID: PMC9760700 DOI: 10.3389/fchem.2022.1076350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/21/2022] [Indexed: 12/11/2022] Open
Abstract
To improve the selective delivery of cisplatin (Cis) to cancer cells, we report and establish the significance of active, targeting drug delivery nanosystems for efficient treatment of lung cancer. Specifically, pH-responsive nano-sized zeolitic imidazolate framework (nZIF-90) was synthesized, post-synthetically modified with an Arg-Gly-Asp peptide motif (RGD@nZIF-90), a known cancer cell homing peptide, and loaded with a large amount of Cis (RGD@Cis⊂nZIF-90). RGD@Cis⊂nZIF-90 was shown to be highly stable under physiological conditions (pH = 7.4) with framework dissociation occurring under slightly acidic conditions (pH = 5.0)-conditions relevant to tumor cells-from which 90% of the encapsulated Cis was released in a sustained manner. In vitro assays demonstrated that RGD@Cis⊂nZIF-90 achieved significantly better cytotoxicity (65% at 6.25 μg ml-1) and selectivity (selectivity index = 4.18 after 48 h of treatment) against adenocarcinoma alveolar epithelial cancer cells (A549) when compared with the unmodified Cis⊂nZIF-90 (22%). Cellular uptake using A549 cells indicated that RGD@Cis⊂nZIF-90 was rapidly internalized leading to significant cell death. After successfully realizing this nanocarrier system, we demonstrated its efficacy in transporting and delivering Cis to cancer cells.
Collapse
Affiliation(s)
- Adamu Abubakar
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Seri Kembangan, Malaysia,Department of Chemical Sciences, Taraba State University, P.M.B, Taraba State, Jalingo, Nigeria
| | - Emilia Abdulmalek
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Seri Kembangan, Malaysia,*Correspondence: Emilia Abdulmalek, ; Kyle E. Cordova,
| | | | - Kyle E. Cordova
- Materials Discovery Research Unit, Advanced Research Centre, Royal Scientific Society, Amman, Jordan,Department of Chemistry, Faculty of Science, UPM, Selangor Darul Ehsan, Malaysia,Foundry of Reticular Materials for Sustainability (FORMS), Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology, UPM, Selangor Darul Ehsan, Malaysia,*Correspondence: Emilia Abdulmalek, ; Kyle E. Cordova,
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Seri Kembangan, Malaysia,Department of Chemistry, Faculty of Science, UPM, Selangor Darul Ehsan, Malaysia,Foundry of Reticular Materials for Sustainability (FORMS), Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology, UPM, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
7
|
Acetalated dextran microparticles for the smart delivery of pyraclostrobin to control Sclerotinia diseases. Carbohydr Polym 2022; 291:119576. [DOI: 10.1016/j.carbpol.2022.119576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/06/2022] [Accepted: 05/03/2022] [Indexed: 11/23/2022]
|
8
|
Bakar SAA, Ali AM, Noor SNFM, Hamid SBS, Azhar NA, Mohamad NM, Ahmad NH. Combination of Goniothalamin and Sol-Gel-Derived Bioactive Glass 45S5 Enhances Growth Inhibitory Activity via Apoptosis Induction and Cell Cycle Arrest in Breast Cancer Cells MCF-7. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5653136. [PMID: 35872839 PMCID: PMC9303150 DOI: 10.1155/2022/5653136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/17/2022] [Indexed: 12/24/2022]
Abstract
Background Combination of natural products with chemically synthesised biomaterials as cancer therapy has attracted great interest lately. Hence, this study is aimed at investigating the combined effects of goniothalamin and bioactive glass 45S5 (GTN-BG) and evaluating their anticancer properties on human breast cancer cells MCF-7. Methods The BG 45S5 was prepared using the sol-gel process followed by characterisation using PSA, BET, SEM/EDS, XRD, and FTIR. The effects of GTN-BG on the proliferation of MCF-7 were assessed by MTT, PrestoBlue, and scratch wound assays. The cell cycle analysis, Annexin-FITC assay, and activation of caspase-3/7, caspase-8, and caspase-9 assays were determined to further explore its mechanism of action. Results The synthesised BG 45S5 was classified as a fine powder, having a rough surface, and contains mesopores of 12.6 nm. EDS analysis revealed that silica and calcium elements are the primary components of BG powders. Both crystalline and amorphous structures were detected with 73% and 27% similarity to Na2Ca2(Si2O7) and hydroxyapatite, respectively. The combination of GTN-BG was more potent than GTN in inhibiting the proliferation of MCF-7 cells. G0/G1 and G2/M phases of the cell cycle were arrested by GTN and GTN-BG. The percentage of viable cells in GTN-BG treatment was significantly lower than that in GTN. In terms of activation of initiator caspases for both extrinsic and intrinsic apoptosis pathways, caspase-8 and caspase-9 were found more effective in response to GTN-BG than GTN. Conclusion The anticancer effect of GTN in MCF-7 cells was improved when combined with BG. The findings provide significant insight into the mechanism of GTN-BG against MCF-7 cells, which can potentially be used as a novel anticancer therapeutic approach.
Collapse
Affiliation(s)
- Siti Aishah Abu Bakar
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Pulau Pinang, Malaysia
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut Campus, 22200 Besut, Terengganu Darul Iman, Malaysia
| | - Abdul Manaf Ali
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut Campus, 22200 Besut, Terengganu Darul Iman, Malaysia
| | - Siti Noor Fazliah Mohd Noor
- Department of Dental Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Shahrul Bariyah Sahul Hamid
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Nur Asna Azhar
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Noor Muzamil Mohamad
- Centralised Laboratory Management Center, Universiti Sultan Zainal Abidin, Besut Campus, 22200 Besut, Terengganu Darul Iman, Malaysia
| | - Nor Hazwani Ahmad
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Pulau Pinang, Malaysia
| |
Collapse
|
9
|
Li C, Zeng X, Qiu S, Gu Y, Zhang Y. Nanomedicine for urologic cancers: diagnosis and management. Semin Cancer Biol 2022; 86:463-475. [PMID: 35660001 DOI: 10.1016/j.semcancer.2022.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 02/08/2023]
Abstract
Urologic cancers accounted for more than 2 million new cases and around 0.8 million deaths in 2020. Although surgery, chemotherapy, and radiotherapy, as well as castration for prostate cancer, remain the cornerstones for managing urologic neoplasms, they can result in severe adverse effects, poor patient compliance, and unsatisfactory survival rates, thus, it is essential to develop novel options that enable the early detection of these malignancies, together with providing accurate diagnoses, and more efficient treatment strategies. Nanomedicine represents an emerging approach that can deliver formulations or drugs across traditional biological barriers in the body and be directed to specific cell types within target organs via active targeting or passive targeting, thus, showing potential to improve the management of urologic cancers. In this review, we discussed the most recent updates on the application of nanomedicines in the diagnosis and treatment of urologic cancers, with focus on prostate, bladder and kidney tumors. We also presented the anti-tumor molecular mechanisms of newly designed nanomedicine for treating urologic cancers, mainly including image-guided surgery, chemotherapy, radiotherapy, gene therapy, immunotherapy, and their synergetic therapy. Current studies have demonstrated the potential advantages of nanomedicine over conventional approaches. However, most developments and new findings in this area have not been validated in clinical trials yet, and therefore, efforts shall be made to translate these research insights into clinical practices for urologic cancers.
Collapse
Affiliation(s)
- Chunyang Li
- Biomedical Big Data Center, Kidney Research Institute, West China Hospital, Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- Biomedical Big Data Center, Kidney Research Institute, West China Hospital, Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Shi Qiu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yonghong Gu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, National Clinical Research Center for Geriatrics, Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Alexandrov DV, Korabel N, Currell F, Fedotov S. Dynamics of intracellular clusters of nanoparticles. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00118-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract
Background
Nanoparticles play a crucial role in nanodiagnostics, radiation therapy of cancer, and they are now widely used to effectively deliver drugs to specific sites, targeting whole organs and down to single cells, in a controlled manner. Therapeutic efficiency of nanoparticles greatly depends on their clustering distribution inside cells. Our purpose is to find the cluster density using Smoluchowski’s coagulation equation with injections.
Results
We obtain an exact cluster density of nanoparticles as the steady-state solution of Smoluchowski’s equation describing clustering due to the fusion of endosomes. We also analyze the unsteady cluster distribution and compare it with the experimental data for time evolution of gold nanoparticle clusters in living cells.
Conclusions
We show the steady cluster density is in good agreement with experimental data on gold nanoparticle distribution inside endosomes. We find that for clusters containing between 1 and 20 nanoparticles, the exact cluster density provides a better description of the existing experimental data than the well-known approximate asymptotic power-law distribution $$x^{-3/2}$$
x
-
3
/
2
Collapse
|
11
|
Kumar P, Kaur N, Kumar R, Banerjee P. α ,β-Unsaturated Carbonyls for One-Pot Transition-Metal-Free Access to 3,6-Dihydro-2 H-pyrans. J Org Chem 2022; 87:7167-7178. [PMID: 35579030 DOI: 10.1021/acs.joc.2c00379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An efficient protocol has been developed for accessing mono-, di-, and trisubstituted 3,6-dihydro-2H-pyran derivatives by simply subjecting α,β-unsaturated carbonyls to the carefully optimized Corey-Chaykovsky reaction conditions. The strategy provides selectively substituted dihydropyran derivatives in good to excellent yields with a broad substrate scope under very mild reaction conditions. Easy transformation of the final 3,6-dihydro-2H-pyran to the valued 5,6-dihydro-2H-pyran-2-one and tetrahydro-2H-pyran derivatives expanded the scope of this methodology to diverse oxacycles. Further, the developed strategy also found application in a two-step route to racemic goniothalamin, which is widely studied for its cytotoxic behavior.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Navpreet Kaur
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Rakesh Kumar
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Prabal Banerjee
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
12
|
Stochastic Model of Virus–Endosome Fusion and Endosomal Escape of pH-Responsive Nanoparticles. MATHEMATICS 2022. [DOI: 10.3390/math10030375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this paper, we set up a stochastic model for the dynamics of active Rab5 and Rab7 proteins on the surface of endosomes and the acidification process that govern the virus–endosome fusion and endosomal escape of pH-responsive nanoparticles. We employ a well-known cut-off switch model for Rab5 to Rab7 conversion dynamics and consider two random terms: white Gaussian and Poisson noises with zero mean. We derive the governing equations for the joint probability density function for the endosomal pH, Rab5 and Rab7 proteins. We obtain numerically the marginal density describing random fluctuations of endosomal pH. We calculate the probability of having a pH level inside the endosome below a critical threshold and therefore the percentage of viruses and pH-responsive nanoparticles escaping endosomes. Our results are in good qualitative agreement with experimental data on viral escape.
Collapse
|
13
|
Grigolo TA, Braga CB, Ornelas C, Russowsky D, Ferreira-Silva GA, Ionta M, Pilli RA. Hybrids of 4-hydroxy derivatives of goniothalamin and piplartine bearing a diester or a 1,2,3-triazole linker as antiproliferative agents. Bioorg Chem 2021; 116:105292. [PMID: 34509797 DOI: 10.1016/j.bioorg.2021.105292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/25/2021] [Accepted: 08/17/2021] [Indexed: 12/22/2022]
Abstract
A library of nine hybrids of 4-hydroxygoniothalamin (2), 4-hydroxypiplartine (4), monastrol (5) and oxo-monastrol (6) was prepared via a modular synthetic route with a diester or a 1,2,3-triazole as linkers. The compounds were assayed against a panel of human cancer cell lines, including MCF-7 (breast adenocarcinoma), HeLa (cervical adenocarcinoma), Caco-2 (colorectal adenocarcinoma) and PC3 (prostate adenocarcinoma), as well as against normal breast (MCF10A) and prostate (PNT2) cells. In general, hybrids with an ester linker containing 4-hydroxypiplartine (4) were more potent than the corresponding hybrids with 4-hydroxygoniothalamin (2). On the other hand, compounds presenting the 1,2,3-triazole linker displayed enhanced cytotoxicity and selectivity when compared to their corresponding hybrids with the diester linker. The 4-hydroxypiplartine-based hybrids 12 and 22 displayed high cytotoxicity (IC50 values below 10 μM) against all cancer cells studied, especially in MCF-7 cells with IC50 values of 1.7 ± 0.1 and 1.6 ± 0.9 μM, respectively. Furthermore, the 4-hydroxygoniothalamin-monastrol hybrid (compound 21) and the 4-hydroxypiplartine-oxo-monastrol hybrid (compound 25), both bearing a 1,2,3-triazole linker, displayed high selectivity and potency towards breast cancer cell line (MCF-7 vs. MCF10 cells, selectivity index = 15.8 and 7.1, respectively), while the 4-hydroxypiplartine -4-hydroxymethylgoniothalamin hybrid with a diester linker (compound 33) showed high selectivity towards melanoma cancer cells (selectivity index = 9.6). Antiproliferative and pro-apoptotic potential of compounds 12 and 22 against MCF-7 cancer cells were further investigated. Cell cycle studies revealed increased G2/M population in MCF-7 cultures as well as reduced G0/G1 population compared to the control groups indicating cell cycle arrest in G2/M phase. In addition, the frequency of positive cells for annexin V was higher in treated samples suggesting that compounds 12 and 22 induce apoptosis in estrogen-positive MCF-7 cells.
Collapse
Affiliation(s)
- Thiago A Grigolo
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970 Campinas, Sao Paulo, Brazil
| | - Carolyne B Braga
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970 Campinas, Sao Paulo, Brazil
| | - Catia Ornelas
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970 Campinas, Sao Paulo, Brazil
| | - Dennis Russowsky
- Institute of Chemistry, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme A Ferreira-Silva
- Institute of Biomedical Sciences, Federal University of Alfenas, UNIFAL-MG, 37130-001 Alfenas, Minas Gerais, Brazil
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Alfenas, UNIFAL-MG, 37130-001 Alfenas, Minas Gerais, Brazil
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970 Campinas, Sao Paulo, Brazil.
| |
Collapse
|
14
|
Wang S, Fontana F, Shahbazi MA, Santos HA. Acetalated dextran based nano- and microparticles: synthesis, fabrication, and therapeutic applications. Chem Commun (Camb) 2021; 57:4212-4229. [PMID: 33913978 DOI: 10.1039/d1cc00811k] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acetalated dextran (Ac-DEX) is a pH-responsive dextran derivative polymer. Prepared by a simple acetalation reaction, Ac-DEX has tunable acid-triggered release profile. Despite its relatively short research history, Ac-DEX has shown great potential in various therapeutic applications. Furthermore, the recent functionalization of Ac-DEX makes versatile derivatives with additional properties. Herein, we summarize the cutting-edge development of Ac-DEX and related polymers. Specifically, we focus on the chemical synthesis, nano- and micro-particle fabrication techniques, the controlled-release mechanisms, and the rational design Ac-DEX-based of drug delivery systems in various biomedical applications. Finally, we briefly discuss the challenges and future perspectives in the field.
Collapse
Affiliation(s)
- Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran and Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
15
|
Kamal NAMA, Abdulmalek E, Fakurazi S, Cordova KE, Abdul Rahman MB. Surface peptide functionalization of zeolitic imidazolate framework-8 for autonomous homing and enhanced delivery of chemotherapeutic agent to lung tumor cells. Dalton Trans 2021; 50:2375-2386. [PMID: 33555001 DOI: 10.1039/d1dt00116g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chemotherapeutic agents used in treating certain cancer types operate in a non-selective manner tending to accumulate in normal, healthy tissue when high doses are used. To mitigate the toxicity effect resulting from this, there is an urgent need to develop active nano delivery systems capable of regulating optimal doses specifically to cancer cells without harming adjacent normal cells. Herein, we report a versatile nanoparticle - zeolitic imidazolate framework-8 (nZIF-8) - that is loaded with a chemotherapeutic agent (gemcitabine; GEM) and surface-functionalized with an autonomous homing system (Arg-Gly-Asp peptide ligand; RGD) via a straightforward, one-pot solvothermal reaction. Successful functionalization of the surface of nZIF-8 loaded GEM (GEM⊂nZIF-8) with RGD was proven by spectroscopic and electron microscopy techniques. This surface-functionalized nanoparticle (GEM⊂RGD@nZIF-8) exhibited enhanced uptake in human lung cancer cells (A549), compared with non-functionalized GEM⊂nZIF-8. The GEM⊂RGD@nZIF-8, experienced not only efficient uptake within A549, but also induced obvious cytotoxicity (75% at a concentration of 10 μg mL-1) and apoptosis (62%) after 48 h treatment when compared to the nanoparticle absent of the RGD homing system (GEM⊂nZIF-8). Most importantly, this surface-functionalized nanoparticle was more selective towards lung cancer cells (A549) than normal human lung fibroblast cells (MRC-5) with a selectivity index (SI) of 3.98. This work demonstrates a new one-pot strategy for realizing a surface-functionalized zeolitic imidazolate framework that actively targets cancer cells via an autonomous homing peptide system to deliver a chemotherapeutic payload effectively.
Collapse
Affiliation(s)
- Nurul Akmarina Mohd Abdul Kamal
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia. and UPM-MAKNA Cancer Laboratory, Institute of Bioscience, UPM, Serdang 43400, Selangor, Malaysia and Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang (UMP), Pekan 26600, Pahang, Malaysia
| | - Emilia Abdulmalek
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia.
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, UPM, Serdang 43400, Selangor, Malaysia
| | - Kyle E Cordova
- Materials Discovery Research Unit, Advanced Research Centre, Royal Scientific Society, Amman 11941, Jordan.
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia. and UPM-MAKNA Cancer Laboratory, Institute of Bioscience, UPM, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
16
|
de Souza KFS, Tófoli D, Pereira IC, Filippin KJ, Guerrero ATG, Paredes-Gamero EJ, de Fatima Cepa Matos M, Garcez WS, Garcez FR, Perdomo RT. A styrylpyrone dimer isolated from Aniba heringeri causes apoptosis in MDA-MB-231 triple-negative breast cancer cells. Bioorg Med Chem 2021; 32:115994. [PMID: 33477019 DOI: 10.1016/j.bmc.2021.115994] [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: 10/31/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 01/11/2023]
Abstract
The styrylpyrone dehydrogoniothalamin (1) and two of its dimers (2 and 3) were isolated from the leaves of Aniba heringeri (Lauraceae). Compound 3 is new, while 1 and 2 are being reported for the first time in this species. Structures were determined by 1D- and 2D-NMR spectroscopy, mass spectrometry, and optical rotation data. Cytotoxic effects and selectivity indices were evaluated in five neoplastic cell lines-PC-3 (prostate), 786-0 (renal), HT-29 (colon), MCF-7, and MDA-MB-231 (breast)-and a non-neoplastic cell line, (NIH/3T3, murine fibroblast). Compound 1 inhibited cell growth by 50% (GI50) at concentrations in the 90.4-175.7 μM range, while 2 proved active against MCF-7 and MDA-MB-231 breast cells (GI50 = 12.24, and 34.22 μM, respectively). Compound 3 showed strong cytotoxicity (GI50 = 4.4 μM) against MDA-MB-231 (an established basal triple-negative breast carcinoma (TNBC) cell line), with a high selective index of 35. This compound was subsequently evaluated for apoptosis induction in MDA-MB-231 cells, using GI50 and 50% lethal concentrations (LC50). Flow cytometry analysis showed that at LC50 compound 3 induced cell death with phosphatidylserine externalization and caspase-3 activation. Apoptotic genes were measured by RT-qPCR, revealing an upregulation of BAX, with an increase in expression of the BAX/BCL2 ratio in treated cells. Fluorescence microscopy disclosed morphological changes related to apoptosis. Overall, these findings showed compound 3 to be a promising prototype against TNBC cells that tend to respond poorly to conventional therapies.
Collapse
Affiliation(s)
- Kamylla Fernanda Souza de Souza
- Laboratory of Molecular Biology and Cell Culture, School of Pharmaceutical Sciences, Food Technology, and Nutrition, Universidade Federal de Mato Grosso do Sul, Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Danilo Tófoli
- Institute of Chemistry, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Indiara Correia Pereira
- Laboratory of Molecular Biology and Cell Culture, School of Pharmaceutical Sciences, Food Technology, and Nutrition, Universidade Federal de Mato Grosso do Sul, Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Kelly Juliana Filippin
- Laboratory of Molecular Biology and Cell Culture, School of Pharmaceutical Sciences, Food Technology, and Nutrition, Universidade Federal de Mato Grosso do Sul, Mato Grosso do Sul, Campo Grande, MS, Brazil
| | | | - Edgar Julian Paredes-Gamero
- Laboratory of Molecular Biology and Cell Culture, School of Pharmaceutical Sciences, Food Technology, and Nutrition, Universidade Federal de Mato Grosso do Sul, Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Maria de Fatima Cepa Matos
- Laboratory of Molecular Biology and Cell Culture, School of Pharmaceutical Sciences, Food Technology, and Nutrition, Universidade Federal de Mato Grosso do Sul, Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Walmir Silva Garcez
- Institute of Chemistry, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | | | - Renata Trentin Perdomo
- Laboratory of Molecular Biology and Cell Culture, School of Pharmaceutical Sciences, Food Technology, and Nutrition, Universidade Federal de Mato Grosso do Sul, Mato Grosso do Sul, Campo Grande, MS, Brazil.
| |
Collapse
|
17
|
Abdelaal MR, Soror SH, Elnagar MR, Haffez H. Revealing the Potential Application of EC-Synthetic Retinoid Analogues in Anticancer Therapy. Molecules 2021; 26:506. [PMID: 33477997 PMCID: PMC7835894 DOI: 10.3390/molecules26020506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background and Aim: All-trans retinoic acid (ATRA) induces differentiation and inhibits growth of many cancer cells. However, resistance develops rapidly prompting the urgent need for new synthetic and potent derivatives. EC19 and EC23 are two synthetic retinoids with potent stem cell neuro-differentiation activity. Here, these compounds were screened for their in vitro antiproliferative and cytotoxic activity using an array of different cancer cell lines. (2) Methods: MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, AV/PI (annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI)), cell cycle analysis, immunocytochemistry, gene expression analysis, Western blotting, measurement of glutamate and total antioxidant concentrations were recruited. (3) Results: HepG2, Caco-2, and MCF-7 were the most sensitive cell lines; HepG2 (ATRA; 36.2, EC19; 42.2 and EC23; 0.74 µM), Caco-2 (ATRA; 58.0, EC19; 10.8 and EC23; 14.7 µM) and MCF-7 (ATRA; 99.0, EC19; 9.4 and EC23; 5.56 µM). Caco-2 cells were selected for further biochemical investigations. Isobologram analysis revealed the combined synergistic effects with 5-fluorouracil with substantial reduction in IC50. All retinoids induced apoptosis but EC19 had higher potency, with significant cell cycle arrest at subG0-G1, -S and G2/M phases, than ATRA and EC23. Moreover, EC19 reduced cellular metastasis in a transwell invasion assay due to overexpression of E-cadherin, retinoic acid-induced 2 (RAI2) and Werner (WRN) genes. (4) Conclusion: The present study suggests that EC-synthetic retinoids, particularly EC19, can be effective, alone or in combinations, for potential anticancer activity to colorectal cancer. Further in vivo studies are recommended to pave the way for clinical applications.
Collapse
Affiliation(s)
- Mohamed R. Abdelaal
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Sameh H. Soror
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Mohamed R. Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt;
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| |
Collapse
|