1
|
Jackson Cullison SR, Flemming JP, Karagoz K, Wermuth PJ, Mahoney MG. Mechanisms of extracellular vesicle uptake and implications for the design of cancer therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70017. [PMID: 39483807 PMCID: PMC11522837 DOI: 10.1002/jex2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024]
Abstract
The translation of pre-clinical anti-cancer therapies to regulatory approval has been promising, but slower than hoped. While innovative and effective treatments continue to achieve or seek approval, setbacks are often attributed to a lack of efficacy, failure to achieve clinical endpoints, and dose-limiting toxicities. Successful efforts have been characterized by the development of therapeutics designed to specifically deliver optimal and effective dosing to tumour cells while minimizing off-target toxicity. Much effort has been devoted to the rational design and application of synthetic nanoparticles to serve as targeted therapeutic delivery vehicles. Several challenges to the successful application of this modality as delivery vehicles include the induction of a protracted immune response that results in their rapid systemic clearance, manufacturing cost, lack of stability, and their biocompatibility. Extracellular vesicles (EVs) are a heterogeneous class of endogenous biologically produced lipid bilayer nanoparticles that mediate intercellular communication by carrying bioactive macromolecules capable of modifying cellular phenotypes to local and distant cells. By genetic, chemical, or metabolic methods, extracellular vesicles (EVs) can be engineered to display targeting moieties on their surface while transporting specific cargo to modulate pathological processes following uptake by target cell populations. This review will survey the types of EVs, their composition and cargoes, strategies employed to increase their targeting, uptake, and cargo release, and their potential as targeted anti-cancer therapeutic delivery vehicles.
Collapse
Affiliation(s)
| | - Joseph P. Flemming
- Rowan‐Virtua School of Osteopathic MedicineRowan UniversityStratfordNew JerseyUSA
| | - Kubra Karagoz
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Mỹ G. Mahoney
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Otolaryngology – Head and Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Ilovaisky AI, Scherbakov AM, Chernoburova EI, Shchetinina MA, Merkulova VM, Bogdanov FB, Sorokin DV, Salnikova DI, Bozhenko EI, Zavarzin IV, Terent'ev AO. Secosteroid diacylhydrazines as novel effective agents against hormone-dependent breast cancer cells. J Steroid Biochem Mol Biol 2024; 244:106597. [PMID: 39127416 DOI: 10.1016/j.jsbmb.2024.106597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
This research aimed to develop novel selective secosteroids that are highly active against hormone-dependent breast cancer. A simple and convenient approach to N'-acylated 13,17-secoestra-1,3,5(10)-trien-17-oic acid hydrazides was disclosed and these novel types of secosteroids were screened for cytotoxicity against the hormone-dependent human breast cancer cell line MCF7. Most secosteroid N'-benzoyl hydrazides have demonstrated high cytotoxicity against MCF7 cells with IC50 values below 5 μM, which are superior to that of the reference drug cisplatin. Hit compounds 2c, 2e and 2i were characterized by high cytotoxicity (IC50 = 1.6-1.9 μM) and very good selectivity towards MCF7 breast cancer cells. The lead secosteroids 2c, 2e and 2i also exhibit antiestrogenic effects and alter the expression of cell cycle regulating proteins. The effect of selected compounds on PARP (poly(ADP-ribose) polymerase) and Bcl-2 (B-cell CLL/lymphoma 2) indicates their proapoptotic potential. The synthesized secosteroids may be considered as new promising anti-breast cancer agents targeting ERα and apoptosis pathways.
Collapse
Affiliation(s)
- Alexey I Ilovaisky
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander M Scherbakov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia; Gause Institute of New Antibiotics, Bol'shaya Pirogovskaya ulitsa 11, Moscow 119021, Russia
| | - Elena I Chernoburova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Marina A Shchetinina
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Valentina M Merkulova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Fedor B Bogdanov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Danila V Sorokin
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Diana I Salnikova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia; N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Eugene I Bozhenko
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Igor V Zavarzin
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander O Terent'ev
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia.
| |
Collapse
|
3
|
Kamle M, Pandhi S, Mishra S, Barua S, Kurian A, Mahato DK, Rasane P, Büsselberg D, Kumar P, Calina D, Sharifi-Rad J. Camptothecin and its derivatives: Advancements, mechanisms and clinical potential in cancer therapy. Med Oncol 2024; 41:263. [PMID: 39382779 DOI: 10.1007/s12032-024-02527-x] [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: 07/30/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024]
Abstract
Camptothecin (CPT), an alkaloid isolated from the Camptotheca tree, has demonstrated significant anticancer properties in a range of malignancies. However, its therapeutic efficacy is limited by its hydrophobicity, poor bioavailability, and systemic toxicity. Derivatives, analogues, and nanoformulations of CPT have been synthesized to overcome these limitations. The aim of this review is to comprehensively analyze existing studies to evaluate the therapeutic efficacy, mechanistic aspects, and clinical potential of CPT and its modified forms, including derivatives, analogues, and nanoformulations, in cancer treatment. A comprehensive literature review was performed using PubMed/Medline, Scopus, and Web of Science databases; articles were selected based on specific inclusion criteria, and data were extracted on the pharmacological profile, clinical studies, and therapeutic efficacy of CPT and its different forms. Current evidence suggests that derivatives and analogues of CPT have improved water solubility, bioavailability, and reduced systemic toxicity compared to CPT. Nanoformulations further enhance targeted delivery and reduce off-target effects. Clinical trials indicate promising outcomes with enhanced survival rates and lower side effects. CPT and its modified forms hold significant promise as potent anticancer agents. Ongoing research and clinical trials are essential for establishing their long-term efficacy and safety; the evidence overwhelmingly supports further development and clinical testing of these compounds.
Collapse
Affiliation(s)
- Madhu Kamle
- Applied Microbiology Laboratory, Department of Forestry, North Eastern Regional Institute of Science and Technology, Nirjuli, India
| | - Shikha Pandhi
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Sadhna Mishra
- Faculty of Agricultural Sciences, GLA University, Mathura, 281406, India
| | - Sreejani Barua
- Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Anju Kurian
- Department of Post Graduate Studies and Research in Food Science, St. Aloysius College (Autonomous), Mangalore, 575003, India
| | - Dipendra Kumar Mahato
- CASS Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Prasad Rasane
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, 144411, India
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Pradeep Kumar
- Department of Botany, University of Lucknow, Uttar Pradesh, Lucknow, India.
- College of Life Science & Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
4
|
Fotopoulos I, Hadjipavlou-Litina D. Approaches for the discovery of cinnamic acid derivatives with anticancer potential. Expert Opin Drug Discov 2024; 19:1281-1291. [PMID: 39105559 DOI: 10.1080/17460441.2024.2387122] [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: 05/20/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Cinnamic acid is a privileged scaffold for the design of biologically active compounds with putative anticancer potential, following different synthetic methodologies and procedures. Since there is a need for the production of potent anticancer, cinnamate moiety can significantly contribute in the design of new and more active anticancer agents. AREAS COVERED In this review, the authors provide a review on the synthetic approaches for the discovery of cinnamic acid derivatives with anticancer potential. Results from molecular simulations, hybridization, and chemical derivatization along with biological experiments in vitro and structural activity relationships are given, described, and discussed by the authors. Information for the mechanism of action is taken from original literature sources. EXPERT OPINION The authors suggest that (i) numerous areas of biology-pharmacology need to be considered: selectivity, in vivo studies, toxicity and drug-likeness, the mechanism of action in animals and humans, development of more efficient assays for various cancer types; (ii) hybridization techniques outbalance in the discovery and production of compounds with higher activity and greater selectivity; (iii) repositioning offers new anticancer cinnamic agents.
Collapse
Affiliation(s)
- Ioannis Fotopoulos
- Department of Pharmaceutical Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | |
Collapse
|
5
|
Andrés CMC, Pérez de la Lastra JM, Munguira EB, Andrés Juan C, Pérez-Lebeña E. Dual-Action Therapeutics: DNA Alkylation and Antimicrobial Peptides for Cancer Therapy. Cancers (Basel) 2024; 16:3123. [PMID: 39335095 PMCID: PMC11429518 DOI: 10.3390/cancers16183123] [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: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer remains one of the most difficult diseases to treat, requiring continuous research into innovative therapeutic strategies. Conventional treatments such as chemotherapy and radiotherapy are effective to a certain extent but often have significant side effects and carry the risk of resistance. In recent years, the concept of dual-acting therapeutics has attracted considerable attention, particularly the combination of DNA alkylating agents and antimicrobial peptides. DNA alkylation, a well-known mechanism in cancer therapy, involves the attachment of alkyl groups to DNA, leading to DNA damage and subsequent cell death. Antimicrobial peptides, on the other hand, have been shown to be effective anticancer agents due to their ability to selectively disrupt cancer cell membranes and modulate immune responses. This review aims to explore the synergistic potential of these two therapeutic modalities. It examines their mechanisms of action, current research findings, and the promise they offer to improve the efficacy and specificity of cancer treatments. By combining the cytotoxic power of DNA alkylation with the unique properties of antimicrobial peptides, dual-action therapeutics may offer a new and more effective approach to fighting cancer.
Collapse
Affiliation(s)
- Celia María Curieses Andrés
- Hospital Clínico Universitario de Valladolid, Avenida de Ramón y Cajal, 3, 47003 Valladolid, Spain; (C.M.C.A.); (E.B.M.)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | - Elena Bustamante Munguira
- Hospital Clínico Universitario de Valladolid, Avenida de Ramón y Cajal, 3, 47003 Valladolid, Spain; (C.M.C.A.); (E.B.M.)
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain;
| | | |
Collapse
|
6
|
Szymczuk D, Markiewicz KH, Niemirowicz-Laskowska K, Sadowska A, Wołosewicz J, Car H, Wilczewska AZ. Magnetic particles with polymeric shells bearing lithocholic and folic acid moieties: Nano-warriors to fight colon cancer. Int J Pharm 2024; 662:124503. [PMID: 39043284 DOI: 10.1016/j.ijpharm.2024.124503] [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: 04/26/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
In the study, we aimed to investigate the activity of nanoformulations containing 5-fluorouracil and polymer-magnetic hybrids bearing membrane-penetrating and ligand-receptor-recognizing agents against colorectal cancer cells. The formation and characterization of iron oxide particles covered with polymeric shells comprising lithocholic acid and folic acid moieties are presented. The efficiency of nanoformulations combined by the simple mixing of low doses of 5-fluorouracil with the obtained hybrids was demonstrated against DLD-1 and HT-29 colon cancer cells. The most pronounced cytotoxic potential against HT-29 cells was observed in the cases of particles based on block and randomly arranged copolymers functionalized by FA motifs with depletion of viable cells by approximately 50 % compared to control cells and cells treated by 5-FU applied in free form. In the case of the DLD-1 cell line, the percentage of viable DLD-1 cells decreased by about 30 to 40% after treatment with the block and randomly arranged copolymer decorated by FA-moiety, when compared to 5-FU at the free form and the untreated control. The induction of apoptosis associated with PS-translocation was determined to be the main mechanism of their cytotoxic effects. Moreover, the safety profiles of the nanoformulations were established through hemolysis assay and the analysis of the viability of human colorectal fibroblasts. It was indicated that all tested nanoparticles met the compatibility requirements at the in vitro level. It should be emphasized that in many cases, there was a significant improvement in the compatibility of hybrids with the FA motif compared to non-functionalized hybrids with the addition of 5-FU. These findings suggest that the presence of FA might modulate the toxicity of chemotherapeutic agents.
Collapse
Affiliation(s)
- Dawid Szymczuk
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland; Doctoral School of Exact and Natural Sciences, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Karolina H Markiewicz
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland.
| | | | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland
| | - Joanna Wołosewicz
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland
| | - Agnieszka Z Wilczewska
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland.
| |
Collapse
|
7
|
Delgado JF, Negussie AH, Varble NA, Mikhail AS, Arrichiello A, Borde T, Saccenti L, Bakhutashvili I, Owen JW, Morhard R, Karanian JW, Pritchard WF, Wood BJ. In vivo Imaging and Pharmacokinetics of Percutaneously Injected Ultrasound and X-ray Imageable Thermosensitive Hydrogel loaded with Doxorubicin versus Free Drug in Swine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.01.610710. [PMID: 39282453 PMCID: PMC11398325 DOI: 10.1101/2024.09.01.610710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Intratumoral injections often lack visibility, leading to unpredictable outcomes such as incomplete tumor coverage, off-target drug delivery and systemic toxicities. This study investigated an ultrasound (US) and x-ray imageable thermosensitive hydrogel based on poloxamer 407 (POL) percutaneously delivered in a healthy swine model. The primary objective was to assess the 2D and 3D distribution of the hydrogel within tissue across three different needle devices and injection sites: liver, kidney, and intercostal muscle region. Secondly, pharmacokinetics of POL loaded with doxorubicin (POLDOX) were evaluated and compared to free doxorubicin injection (DOXSoln) with a Single End Hole Needle. Utilizing 2D and 3D morphometrics from US and x-ray imaging techniques such as Computed Tomography (CT) and Cone Beam CT (CBCT), we monitored the localization and leakage of POLDOX over time. Relative iodine concentrations measured with CBCT following incorporation of an iodinated contrast agent in POL indicated potential drug diffusion and advection transport. Furthermore, US imaging revealed temporal changes, suggesting variations in acoustic intensity, heterogeneity, and echotextures. Notably, 3D reconstruction of the distribution of POL and POLDOX from 2D ultrasound frames was achieved and morphometric data obtained. Pharmacokinetic analysis revealed lower systemic exposure of the drug in various organs with POLDOX formulation compared to DOXSoln formulation. This was demonstrated by a lower area under the curve (852.1 ± 409.1 ng/mL·h vs 2283.4 ± 377.2 ng/mL·h) in the plasma profile, suggesting a potential reduction in systemic toxicity. Overall, the use of POL formulation offers a promising strategy for precise and localized drug delivery, that may minimize adverse effects. Dual modality POL imaging enabled analysis of patterns of gel distribution and morphology, alongside of pharmacokinetics of local delivery. Incorporating hydrogels into drug delivery systems holds significant promise for improving the predictability of the delivered drug and enhancing spatial conformability. These advancements can potentially enhance the safety and precision of anticancer therapy.
Collapse
Affiliation(s)
- Jose F. Delgado
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD
| | - Ayele H. Negussie
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Nicole A. Varble
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
- Philips Healthcare, Cambridge, MA
| | - Andrew S. Mikhail
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Antonio Arrichiello
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
- UOS of Interventional Radiology, Department of Diagnostic and Interventional Radiology, Ospedale Maggiore di Lodi, Largo Donatori del Sangue, Lodi, Italy
| | - Tabea Borde
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Laetitia Saccenti
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Ivane Bakhutashvili
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Joshua W. Owen
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Robert Morhard
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - John W. Karanian
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - William F. Pritchard
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Bradford J. Wood
- Center for Interventional Oncology, Clinical Center, National Institutes of Health, Bethesda, MD
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD
| |
Collapse
|
8
|
Li C, Shi K, Zhao S, Liu J, Zhai Q, Hou X, Xu J, Wang X, Liu J, Wu X, Fan W. Natural-source payloads used in the conjugated drugs architecture for cancer therapy: Recent advances and future directions. Pharmacol Res 2024; 207:107341. [PMID: 39134188 DOI: 10.1016/j.phrs.2024.107341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024]
Abstract
Drug conjugates are obtained from tumor-located vectors connected to cytotoxic agents via linkers, which are designed to deliver hyper-toxic payloads directly to targeted cancer cells. These drug conjugates include antibody-drug conjugates (ADCs), peptide-drug conjugates (PDCs), small molecule-drug conjugates (SMDCs), nucleic acid aptamer-drug conjugates (ApDCs), and virus-like drug conjugate (VDCs), which show great therapeutic value in the clinic. Drug conjugates consist of a targeting carrier, a linker, and a payload. Payloads are key therapy components. Cytotoxic molecules and their derivatives derived from natural products are commonly used in the payload portion of conjugates. The ideal payload should have sufficient toxicity, stability, coupling sites, and the ability to be released under specific conditions to kill tumor cells. Microtubule protein inhibitors, DNA damage agents, and RNA inhibitors are common cytotoxic molecules. Among these conjugates, cytotoxic molecules of natural origin are summarized based on their mechanism of action, conformational relationships, and the discovery of new derivatives. This paper also mentions some cytotoxic molecules that have the potential to be payloads. It also summarizes the latest technologies and novel conjugates developed in recent years to overcome the shortcomings of ADCs, PDCs, SMDCs, ApDCs, and VDCs. In addition, this paper summarizes the clinical trials conducted on conjugates of these cytotoxic molecules over the last five years. It provides a reference for designing and developing safer and more efficient conjugates.
Collapse
Affiliation(s)
- Cuiping Li
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Kourong Shi
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Siyuan Zhao
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Juan Liu
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Qiaoli Zhai
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Xiaoli Hou
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Jie Xu
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Xinyu Wang
- Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Jiahui Liu
- Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China.
| | - Xin Wu
- Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China; Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Wei Fan
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| |
Collapse
|
9
|
Nabi SA, Ramzan F, Lone MS, Nainwal LM, Hamid A, Batool F, Husain M, Samim M, Shafi S, Sharma K, Bano S, Javed K. Halogen substituted aurones as potential apoptotic agents: synthesis, anticancer evaluation, molecular docking, ADMET and DFT study. J Biomol Struct Dyn 2024; 42:7610-7627. [PMID: 37517055 DOI: 10.1080/07391102.2023.2240897] [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: 02/03/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023]
Abstract
A series of halogen-substituted aurone derivatives (2a-k) were synthesized and evaluated for an anti-proliferative study against NCI 60 cancer cell line panel and showed that most of the compounds predominantly exhibited promising activity against MCF-7. Compound 2e exhibited promising anticancer activity against the MCF-7 cancer cell line with 84.98% percentage growth inhibition in a single dose assay of 10 μM with an IC50 value of 8.157 ± 0.713 μM. In apoptotic assay, the effect of compound 2e on the cell cycle progression indicated that exposure of MCF-7 cells to compound 2e induced a significant disruption in the cell cycle profile including a time-dependent decrease in the cell population at G0/G1 and G2/M phase and arrests the cell cycle at the S phase. In silico, molecular docking ADME and toxicity studies of all compounds were also carried out. The docking study revealed that all the aurone derivatives displayed good docking scores ranging from -7.066 to -8.573. The results of Molecular Electrostatic Potential Mapping (MESP) and Density Functional Theory (DFT) studies of the most active compound 2e and least active compound 2k also favoured the experimental results.
Collapse
Affiliation(s)
- Syed Ayaz Nabi
- Department of Chemistry, School of Chemical and Life Sciences, New Delhi, India
| | - Farhat Ramzan
- Department of Chemistry, School of Chemical and Life Sciences, New Delhi, India
| | - Mehak Saba Lone
- Department of Chemistry, School of Chemical and Life Sciences, New Delhi, India
| | - Lalit Mohan Nainwal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, New Delhi, India
- Department of Pharmacy, School of Medical & Allied Sciences, G. D. Goenka University, Gurugram, Haryana, India
| | - Aabid Hamid
- Department of Chemistry, Birla Institute of Technology and Science Pilani, Pilani, Rajasthan, India
- Theoretical Chemistry Section, Chemistry Division, Bhabha Atomic Research Centre, Mumbai, India
| | | | | | - Mohammed Samim
- Department of Chemistry, School of Chemical and Life Sciences, New Delhi, India
| | - Syed Shafi
- Department of Chemistry, School of Chemical and Life Sciences, New Delhi, India
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Sameena Bano
- Department of Computer Science and Engineering, School of Engineering Sciences and Technology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Kalim Javed
- Department of Chemistry, School of Chemical and Life Sciences, New Delhi, India
| |
Collapse
|
10
|
Jeong J, Park S, Heo KN, Park SM, Min S, Ah YM, Han JM, Lee JY. Comprehensive analysis of nationwide anticancer drug-related complications in Korea: incidence, types, and cancer-specific considerations in contemporary oncology. Ther Adv Med Oncol 2024; 16:17588359241272970. [PMID: 39206378 PMCID: PMC11350537 DOI: 10.1177/17588359241272970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
Background The rising global incidence of cancer has increased the demand for chemotherapy, which is a crucial treatment modality. Recent advancements in cancer treatment, including targeted agents and immunotherapy, have introduced complications owing to their specific mechanisms. However, comprehensive studies of the combined complications of these approaches are lacking. Objectives This study aimed to comprehensively assess and analyze the overall incidence of anticancer drug-related complications in a nationwide patient cohort, utilizing a customized National Health Insurance Sharing Service database in Korea. Design Retrospective cohort study. Methods We included patients who were prescribed anticancer drugs (excluding endocrine agents) and diagnosed with cancer. For the type of cancer classification, the International Statistical Classification of Diseases and Related Health Problems, Tenth Revision (ICD-10) was used and anticancer drugs were classified based on the Anatomical Therapeutic Chemical code. We classified cancer into 18 types based on the ICD-10 code and delineated cancer-related complications into 12 categories. Complications included hematological, gastrointestinal, infectious, cardiovascular, major bleeding, endocrine, neurotoxic, nephrotoxic, dermatological, pulmonary, musculoskeletal, and hepatotoxic effects. Result We included 294,544 patients diagnosed with cancer and administered anticancer drugs between 2016 and 2018, with follow-up continuing until 2021. We identified 486,929 anticancer drug-related complications, with an incidence of 1843.6 per 1000 person-years (PY). Anemia was the most common complication, with a rate of 763.7 per 1000 PY, followed by febrile neutropenia (295.7) and nausea/vomiting (246.9). Several complications peaked during the first months following the initiation of anticancer drug therapy; however, herpes, skin infection, heart failure, and peripheral neuropathy peaked at 6-12 months. Among major cancers, breast cancer had the lowest overall incidence of complications. Targeted therapies revealed lower complication rates than cytotoxic chemotherapy; however, they also required careful monitoring of rash. Conclusion This study highlights the importance of the proactive management of anticancer drug-related complications for patient care improvement.
Collapse
Affiliation(s)
- Jonghyun Jeong
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Soyoung Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Kyu-Nam Heo
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Soh Mee Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Department of Pharmacy, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sangil Min
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young-Mi Ah
- College of Pharmacy, Yeungnam University, Gyeongsangbuk-do, Republic of Korea
| | - Ji Min Han
- College of Pharmacy, Chungbuk National University, 194-21 Osongsaengmyeong 1-ro, Osong-eup, Cheongju 28160, Republic of Korea
| | - Ju-Yeun Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| |
Collapse
|
11
|
Yoo H, Kim Y, Kim J, Cho H, Kim K. Overcoming Cancer Drug Resistance with Nanoparticle Strategies for Key Protein Inhibition. Molecules 2024; 29:3994. [PMID: 39274842 PMCID: PMC11396748 DOI: 10.3390/molecules29173994] [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: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Drug resistance remains a critical barrier in cancer therapy, diminishing the effectiveness of chemotherapeutic, targeted, and immunotherapeutic agents. Overexpression of proteins such as B-cell lymphoma 2 (Bcl-2), inhibitor of apoptosis proteins (IAPs), protein kinase B (Akt), and P-glycoprotein (P-gp) in various cancers leads to resistance by inhibiting apoptosis, enhancing cell survival, and expelling drugs. Although several inhibitors targeting these proteins have been developed, their clinical use is often hampered by systemic toxicity, poor bioavailability, and resistance development. Nanoparticle-based drug delivery systems present a promising solution by improving drug solubility, stability, and targeted delivery. These systems leverage the Enhanced Permeation and Retention (EPR) effect to accumulate in tumor tissues, reducing off-target toxicity and increasing therapeutic efficacy. Co-encapsulation strategies involving anticancer drugs and resistance inhibitors within nanoparticles have shown potential in achieving coordinated pharmacokinetic and pharmacodynamic profiles. This review discusses the mechanisms of drug resistance, the limitations of current inhibitors, and the advantages of nanoparticle delivery systems in overcoming these challenges. By advancing these technologies, we can enhance treatment outcomes and move towards more effective cancer therapies.
Collapse
Affiliation(s)
- Hyeonji Yoo
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yeonjin Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinseong Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
12
|
Cheong HH, Zuo W, Chen J, Un CW, Si YW, Wong KH, Kwok HF, Siu SWI. Identification of Anticancer Peptides from the Genome of Candida albicans: in Silico Screening, in Vitro and in Vivo Validations. J Chem Inf Model 2024; 64:6174-6189. [PMID: 39008832 DOI: 10.1021/acs.jcim.4c00501] [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: 07/17/2024]
Abstract
Anticancer peptides (ACPs) are promising future therapeutics, but their experimental discovery remains time-consuming and costly. To accelerate the discovery process, we propose a computational screening workflow to identify, filter, and prioritize peptide sequences based on predicted class probability, antitumor activity, and toxicity. The workflow was applied to identify novel ACPs with potent activity against colorectal cancer from the genome sequences of Candida albicans. As a result, four candidates were identified and validated in the HCT116 colon cancer cell line. Among them, PCa1 and PCa2 emerged as the most potent, displaying IC50 values of 3.75 and 56.06 μM, respectively, and demonstrating a 4-fold selectivity for cancer cells over normal cells. In the colon xenograft nude mice model, the administration of both peptides resulted in substantial inhibition of tumor growth without causing significant adverse effects. In conclusion, this work not only contributes a proven computational workflow for ACP discovery but also introduces two peptides, PCa1 and PCa2, as promising candidates poised for further development as targeted therapies for colon cancer. The method as a web service is available at https://app.cbbio.online/acpep/home and the source code at https://github.com/cartercheong/AcPEP_classification.git.
Collapse
Affiliation(s)
- Hong-Hin Cheong
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Weimin Zuo
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Jiarui Chen
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Chon-Wai Un
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Yain-Whar Si
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Koon Ho Wong
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR 999078, China
| | - Shirley W I Siu
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macau SAR 999078, China
- Institute of Science and Environment, University of Saint Joseph, Estrada Marginal da Ilha Verde 14-17, Macau SAR 999078, China
| |
Collapse
|
13
|
Janssen FW, Lak NSM, Janda CY, Kester LA, Meister MT, Merks JHM, van den Heuvel-Eibrink MM, van Noesel MM, Zsiros J, Tytgat GAM, Looijenga LHJ. A comprehensive overview of liquid biopsy applications in pediatric solid tumors. NPJ Precis Oncol 2024; 8:172. [PMID: 39097671 PMCID: PMC11297996 DOI: 10.1038/s41698-024-00657-z] [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: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 08/05/2024] Open
Abstract
Liquid biopsies are emerging as an alternative source for pediatric cancer biomarkers with potential applications during all stages of patient care, from diagnosis to long-term follow-up. While developments within this field are reported, these mainly focus on dedicated items such as a specific liquid biopsy matrix, analyte, and/or single tumor type. To the best of our knowledge, a comprehensive overview is lacking. Here, we review the current state of liquid biopsy research for the most common non-central nervous system pediatric solid tumors. These include neuroblastoma, renal tumors, germ cell tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma and other soft tissue sarcomas, and liver tumors. Within this selection, we discuss the most important or recent studies involving liquid biopsy-based biomarkers, anticipated clinical applications, and the current challenges for success. Furthermore, we provide an overview of liquid biopsy-based biomarker publication output for each tumor type based on a comprehensive literature search between 1989 and 2023. Per study identified, we list the relevant liquid biopsy-based biomarkers, matrices (e.g., peripheral blood, bone marrow, or cerebrospinal fluid), analytes (e.g., circulating cell-free and tumor DNA, microRNAs, and circulating tumor cells), methods (e.g., digital droplet PCR and next-generation sequencing), the involved pediatric patient cohort, and proposed applications. As such, we identified 344 unique publications. Taken together, while the liquid biopsy field in pediatric oncology is still behind adult oncology, potentially relevant publications have increased over the last decade. Importantly, steps towards clinical implementation are rapidly gaining ground, notably through validation of liquid biopsy-based biomarkers in pediatric clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Michael T Meister
- Princess Máxima Center, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Johannes H M Merks
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center, Utrecht, the Netherlands
- Wilhelmina Children's Hospital-Division of CHILDHEALTH, University Medical Center Utrech, University of Utrecht, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | | | - Godelieve A M Tytgat
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Leendert H J Looijenga
- Princess Máxima Center, Utrecht, the Netherlands.
- Department of Pathology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Ghosh S, Das SK, Sinha K, Ghosh B, Sen K, Ghosh N, Sil PC. The Emerging Role of Natural Products in Cancer Treatment. Arch Toxicol 2024; 98:2353-2391. [PMID: 38795134 DOI: 10.1007/s00204-024-03786-3] [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: 02/15/2024] [Accepted: 05/08/2024] [Indexed: 05/27/2024]
Abstract
The exploration of natural products as potential agents for cancer treatment has garnered significant attention in recent years. In this comprehensive review, we delve into the diverse array of natural compounds, including alkaloids, carbohydrates, flavonoids, lignans, polyketides, saponins, tannins, and terpenoids, highlighting their emerging roles in cancer therapy. These compounds, derived from various botanical sources, exhibit a wide range of mechanisms of action, targeting critical pathways involved in cancer progression such as cell proliferation, apoptosis, angiogenesis, and metastasis. Through a meticulous examination of preclinical and clinical studies, we provide insights into the therapeutic potential of these natural products across different cancer types. Furthermore, we discuss the advantages and challenges associated with their use in cancer treatment, emphasizing the need for further research to optimize their efficacy, pharmacokinetics, and delivery methods. Overall, this review underscores the importance of natural products in advancing cancer therapeutics and paves the way for future investigations into their clinical applications.
Collapse
Affiliation(s)
- Sumit Ghosh
- Department of Zoology, Ramakrishna Mission Vidyamandira, Belur Math, Howrah, 711202, India
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India
| | - Sanjib Kumar Das
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India
| | - Krishnendu Sinha
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India.
| | - Biswatosh Ghosh
- Department of Zoology, Bidhannagar College, Kolkata, 700064, India
| | - Koushik Sen
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India
| | - Nabanita Ghosh
- Department of Zoology, Maulana Azad College, Kolkata, 700013, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India.
| |
Collapse
|
15
|
Sun H, Wienkers LC, Lee A. Beyond cytotoxic potency: disposition features required to design ADC payload. Xenobiotica 2024; 54:442-457. [PMID: 39017706 DOI: 10.1080/00498254.2024.2381139] [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: 06/06/2024] [Accepted: 07/13/2024] [Indexed: 07/18/2024]
Abstract
1. Antibody-drug conjugates (ADCs) have demonstrated impressive clinical usefulness in treating several types of cancer, with the notion of widening of the therapeutic index of the cytotoxic payload through the minimisation of the systemic toxicity. Therefore, choosing the most appropriate payload molecule is a particularly important part of the early design phase of ADC development, especially given the highly competitive environment ADCs find themselves in today.2. The focus of the current review is to describe critical attributes/considerations needed in the discovery and ultimately development of cytotoxic payloads in support of ADC design. In addition to potency, several key dispositional characteristics including solubility, permeability and bystander effect, pharmacokinetics, metabolism, and drug-drug interactions, are described as being an integral part of the integrated activities required in the design of clinically safe and useful ADC therapeutic agents.
Collapse
Affiliation(s)
- Hao Sun
- Clinical Pharmacology and Translational Sciences, Pfizer Oncology Division, Pfizer, Inc, Bothell, WA, USA
| | - Larry C Wienkers
- Clinical Pharmacology and Translational Sciences, Pfizer Oncology Division, Pfizer, Inc, Bothell, WA, USA
| | - Anthony Lee
- Clinical Pharmacology and Translational Sciences, Pfizer Oncology Division, Pfizer, Inc, Bothell, WA, USA
| |
Collapse
|
16
|
Sharma D, Dhobi M, Lather V, Pandita D. An insight into the therapeutic effects of isoliquiritigenin in breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03282-6. [PMID: 39007925 DOI: 10.1007/s00210-024-03282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Breast cancer ranks as the most widespread malignant condition in women, emerging as a primary contributor to mortality. The primary challenges in cancer treatments involve undesirable side effects. Therefore, exploring natural compounds as additional therapy could provide valuable insights. Isoliquiritigenin (ILN), an isoflavonoid featuring a chalcone moiety primarily sourced from Glycyrrhiza species, has garnered increasing interest in breast cancer research. This review aims to provide a comprehensive understanding of ILN's mechanisms of action in breast cancer, drawing from a range of in vitro and in vivo studies. ILN primarily acts by inhibiting angiogenesis, aromatase, inflammation, and cell proliferation, and preventing invasion and metastasis. Mechanistically, it downregulates miR-374a, phosphoinositide-3-kinase-protein kinase B/Akt, maternal embryonic leucine zipper kinase, vascular endothelial growth factor, and estrogen receptor protein levels, and causes enhancement of Wnt inhibitory factor-1, and Unc-51-like kinase 1 expression to treat breast cancer. ILN emerges as a promising natural option, offering therapeutic advantages with minimal side effects. However, it is important to note that current research on ILN is primarily limited to preclinical models, underscoring the need for further investigation to validate its potential efficacy.
Collapse
Affiliation(s)
- Divya Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India
| | - Mahaveer Dhobi
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India.
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector 125, Noida, 201313, India.
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR) Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India.
- Centre for Advanced Formulation Technology (CAFT), Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India.
| |
Collapse
|
17
|
Zhang Y, Tang N, Zhou H, Zhu Y. Surface engineered multifunctional nano-systems for localised drug delivery against thyroid cancer: A review of current practices. Biomed Pharmacother 2024; 176:116840. [PMID: 38820975 DOI: 10.1016/j.biopha.2024.116840] [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: 02/09/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Thyroid cancer, the most prevalent cancer of the endocrine system and cervical region, has experienced a significant increase in incidence over recent decades. Nanomedicine has fundamentally revolutionized cancer treatment, particularly through the development of multifunctional nano-therapeutics. The progress in this field has been facilitated by the distinctive properties of nanomaterials, such as their capacity to perform several functions, be modified, and offer various detection methods. These features allow for non-invasive and practical diagnostic techniques through versatile imaging. Surface engineering plays a pivotal role in the design of multifunctional nano-systems for localized drug delivery against thyroid cancer. Nano-systems can be customized via surface modification techniques, such as functionalization with targeting ligands and inclusion of therapeutic drugs. This customization allows the nano-systems to specifically target cancer cells while reducing the impact on non-target cells. As a result, bovine serum albumin-coated nanostructures have emerged as powerful diagnostic and targeting nanosystems for thyroid cancer. This targeted strategy enhances the effectiveness of cancer treatment while reducing overall body toxicity. This comprehensive review aims to provide an extensive overview of the latest advancements in surface-engineered nanoparticle-based approaches for both diagnosing and treating thyroid cancer. It highlights the promising research endeavors aimed at creating novel and effective multifunctional nanomedicine for localized delivery to thyroid cancer sites. The review examines different nanomedicines that have been developed for cancer treatment and diagnosis. It also analyzes the current trends, future possibilities, and obstacles in this rapidly advancing sector. By synthesizing the current state of knowledge on surface-engineered multifunctional nano-systems, this review contributes to a better understanding of their potential applications in thyroid cancer treatment and paves the way for future research directions in this promising field of nanomedicine.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Nie Tang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Hui Zhou
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Ying Zhu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
18
|
Chowdhury R, Bhuia MS, Wilairatana P, Afroz M, Hasan R, Ferdous J, Rakib AI, Sheikh S, Mubarak MS, Islam MT. An insight into the anticancer potentials of lignan arctiin: A comprehensive review of molecular mechanisms. Heliyon 2024; 10:e32899. [PMID: 38988539 PMCID: PMC11234030 DOI: 10.1016/j.heliyon.2024.e32899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 07/12/2024] Open
Abstract
Natural products are being developed as possible treatment options due to the rising prevalence of cancer and the harmful side effects of synthetic medications. Arctiin is a naturally occurring lignan found in numerous plants and exhibits different pharmacological activities, along with cancer. To elucidate the anticancer properties and underlying mechanisms of action, a comprehensive search of various electronic databases was conducted using appropriate keywords to identify relevant publications. The findings suggest that arctiin exhibits anticancer properties against tumor formation and various cancers such as cervical, myeloma, prostate, endothelial, gastric, and colon cancers in several preclinical pharmacological investigations. This naturally occurring compound exerts its anticancer effect through different cellular mechanisms, including mitochondrial dysfunction, cell cycle at different phases (G2/M), inhibition of cell proliferation, apoptotic cell death, and cytotoxic effects, as well as inhibition of migration and invasion of various malignant cells. Moreover, the study also revealed that, among the various cellular pathways, arctiin was shown to be more potent in terms of the PI3K/AKT and JAK/STAT signaling pathways. However, pharmacokinetic investigation indicated the compound's poor oral bioavailability. Because of these findings, arctiin might be considered a promising chemotherapeutic drug candidate.
Collapse
Affiliation(s)
- Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
| | - Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
| | - Rubel Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Asraful Islam Rakib
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
| | - Salehin Sheikh
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Gopalganj 8100, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna 9208, Bangladesh
| |
Collapse
|
19
|
Saberian E, Jenča A, Petrášová A, Zare-Zardini H, Ebrahimifar M. Application of Scaffold-Based Drug Delivery in Oral Cancer Treatment: A Novel Approach. Pharmaceutics 2024; 16:802. [PMID: 38931923 PMCID: PMC11207321 DOI: 10.3390/pharmaceutics16060802] [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: 05/04/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
This comprehensive review consolidates insights from two sources to emphasize the transformative impact of scaffold-based drug delivery systems in revolutionizing oral cancer therapy. By focusing on their core abilities to facilitate targeted and localized drug administration, these systems enhance therapeutic outcomes significantly. Scaffolds, notably those coated with anti-cancer agents such as cisplatin and paclitaxel, have proven effective in inhibiting oral cancer cell proliferation, establishing a promising avenue for site-specific drug delivery. The application of synthetic scaffolds, including Poly Ethylene Glycol (PEG) and poly(lactic-co-glycolic acid) (PLGA), and natural materials, like collagen or silk, in 3D systems has been pivotal for controlled release of therapeutic agents, executing diverse anti-cancer strategies. A key advancement in this field is the advent of smart scaffolds designed for sequential cancer therapy, which strive to refine drug delivery systems, minimizing surgical interventions, accentuating the significance of 3D scaffolds in oral cancer management. These systems, encompassing local drug-coated scaffolds and other scaffold-based platforms, hold the potential to transform oral cancer treatment through precise interventions, yielding improved patient outcomes. Local drug delivery via scaffolds can mitigate systemic side effects typically associated with chemotherapy, such as nausea, alopecia, infections, and gastrointestinal issues. Post-drug release, scaffolds foster a conducive environment for non-cancerous cell growth, adhering and proliferation, demonstrating restorative potential. Strategies for controlled and targeted drug delivery in oral cancer therapy span injectable self-assembling peptide hydrogels, nanocarriers, and dual drug-loaded nanofibrous scaffolds. These systems ensure prolonged release, synergistic effects, and tunable targeting, enhancing drug delivery efficiency while reducing systemic exposure. Smart scaffolds, capable of sequential drug release, transitioning to cell-friendly surfaces, and enabling combinatorial therapy, hold the promise to revolutionize treatment by delivering precise interventions and optimized outcomes. In essence, scaffold-based drug delivery systems, through their varied forms and functionalities, are reshaping oral cancer therapy. They target drug delivery efficiency, diminish side effects, and present avenues for personalization. Challenges like fabrication intricacy, biocompatibility, and scalability call for additional research. Nonetheless, the perspective on scaffold-based systems in oral cancer treatment is optimistic, as ongoing advancements aim to surmount current limitations and fully leverage their potential in cancer therapy.
Collapse
Affiliation(s)
- Elham Saberian
- Klinika and Akadémia Košice, Pavol Jozef Šafárik University, n.o. Bačíkova 7, 04001 Kosice, Slovakia;
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, UPJS LF, Pavol Jozef Šafárik University, n.o. Bačíkova 7, 04001 Kosice, Slovakia; (A.J.); (A.P.)
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, UPJS LF, Pavol Jozef Šafárik University, n.o. Bačíkova 7, 04001 Kosice, Slovakia; (A.J.); (A.P.)
| | - Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod 89616-99557, Iran
| | - Meysam Ebrahimifar
- Department of Toxicity, Faculty of Pharmacy, Islamic Azad University, Shahreza Branch, Shahreza 81796-35875, Iran
| |
Collapse
|
20
|
Cörüt R, Altundağ O, Eyüboğlu F. Incidence and Clinical Characteristics of Drug-Induced Lung Disease Among Chemotherapy Recipients. Cureus 2024; 16:e63408. [PMID: 39077293 PMCID: PMC11283914 DOI: 10.7759/cureus.63408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 07/31/2024] Open
Abstract
Background Chemotherapeutic agents treat cancer and some inflammatory diseases due to their immunosuppressive effects. While effective, these drugs can cause drug-induced lung disease (DILD), a serious adverse effect with limited data regarding its incidence and clinical presentation. Methods This retrospective study included 20 patients diagnosed with DILD out of 1,231 patients treated with chemotherapeutic agents who presented with symptoms such as cough, fever, dyspnea, and chest pain at an oncology outpatient clinic. Patients underwent assessments including clinical examination, chest radiography, high-resolution computed tomography, and, in some cases, video-assisted thoracoscopic surgery. A statistical analysis was performed to determine the incidence and evaluate the clinical characteristics of DILD. Results The incidence of DILD among patients treated with chemotherapeutic agents was 0.27%. The female/male ratio was 11/9, with a mean age of 53.2 years. Common symptoms included cough (70%), dyspnea (60%), fever (50%), and sputum production (40%). Imaging revealed pleural effusion, reticular patterns, and consolidation in varying proportions. Common agents causing pulmonary toxicity included bleomycin, cyclophosphamide, and methotrexate, among others. Importantly, 95% of patients showed improvement with steroid treatment, although statistical significance was not achieved (p > 0.05). Conclusion The findings highlight the need for heightened awareness and monitoring of DILD in patients receiving chemotherapeutic treatments. Early diagnosis and prompt treatment initiation are crucial to managing this potentially severe complication. This study underscores the importance of considering pulmonary risks when prescribing chemotherapeutic agents and provides foundational data for future research.
Collapse
Affiliation(s)
- Ruhsel Cörüt
- Pulmonology, Giresun University Faculty of Medicine, Giresun, TUR
| | - Ozden Altundağ
- Medical Oncology, Faculty of Medicine, Başkent University, Ankara, TUR
| | - Füsun Eyüboğlu
- Pulmonology, Faculty of Medicine, Başkent University, Ankara, TUR
| |
Collapse
|
21
|
Bokhari SS, Ali T, Naeem M, Hussain F, Nasir A. Recent advances in nanoformulation-based delivery for cancer immunotherapy. Nanomedicine (Lond) 2024; 19:1253-1269. [PMID: 38717427 PMCID: PMC11285355 DOI: 10.1080/17435889.2024.2343273] [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: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 07/25/2024] Open
Abstract
Cancer is one of the leading causes of mortality worldwide, and its treatment faces several challenges. Phytoconstituents derived from recently discovered medicinal plants through nanotechnology potentially target cancer cells via PI3K/Akt/mTOR pathways and exert their effects selectively through the generation of reactive oxygen species through β-catenin inhibition, DNA damage, and increasing caspase 3/9 and p53 expression. These nanocarriers act specifically against different cancer cell lines such as HT-29, MOLT-4 human leukemia cancer and MCF-7 cell lines SKOV-3, Caov-3, SW-626, HepG2, A-549, HeLa, and MCF-7. This review comprehensively elaborates on the cellular and molecular mechanisms, and therapeutic prospects of various plant-mediated nanoformulations to attain a revolutionary shift in cancer immunotherapy.
Collapse
Affiliation(s)
- Seyedeh Saimeh Bokhari
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Tayyab Ali
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Fatma Hussain
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Abdul Nasir
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| |
Collapse
|
22
|
Shahbaz S, Esmaeili M, Fathian Nasab MH, Imani Z, Bafkary R, Amini M, Atyabi F, Dinarvand R. PEGylated mesoporous silica core-shell redox-responsive nanoparticles for delivering paclitaxel to breast cancer cells. Int J Pharm 2024; 655:124024. [PMID: 38537920 DOI: 10.1016/j.ijpharm.2024.124024] [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: 01/21/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
Controlling the drug release and restricting its presence in healthy organs is extremely valuable. In this study, mesoporous silica nanoparticles (MSN) as the core, loaded with paclitaxel (PTX), were coated with a non-porous silica shell functionalized with disulfide bonds. The nanoparticles were further coated with polyethylene glycol (PEG) via disulfide linkages. We analyzed the physicochemical properties of nanoparticles, including hydrodynamic size via Dynamic Light Scattering (DLS), zeta potential, X-ray Diffraction (XRD) patterns, Fourier-Transform Infrared (FTIR) spectra, and imaging through Transmission Electron Microscopy (TEM) and Scanning Electron Microscopy (SEM). The drug release profile in two distinct glutathione (GSH) concentrations of 2 µM and 10 µM was measured. The cellular uptake of nanoparticles by MCF-7 cell line was determined using Confocal Laser Scanning Microscopy (CLSM) images and flow cytometry. Furthermore, the cell viability and the capability of nanoparticles to induce apoptosis in MCF-7 cell line were studied using the MTT assay and flow cytometry, respectively. Our investigations revealed that the release of PTX from the drug delivery system was redox-responsive. Also, results indicated an elevated level of cellular uptake and efficient induction of apoptosis, underscoring the promising potential of this redox-responsive drug delivery system for breast cancer therapy.
Collapse
Affiliation(s)
- Saeed Shahbaz
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahta Esmaeili
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zhila Imani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Bafkary
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Leicester School of Pharmacy, Leicester Institute for Pharmaceutical Innovation, De Montfort University, Leicester, UK.
| |
Collapse
|
23
|
Vafa RG, Sabahizadeh A, Mofarrah R. Guarding the heart: How SGLT-2 inhibitors protect against chemotherapy-induced cardiotoxicity: SGLT-2 inhibitors and chemotherapy-induced cardiotoxicity. Curr Probl Cardiol 2024; 49:102350. [PMID: 38128634 DOI: 10.1016/j.cpcardiol.2023.102350] [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: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
The introduction of chemotherapy agents has significantly transformed cancer treatment, with anthracyclines being one of the most commonly used drugs. While these agents have proven to be highly effective against various types of cancers, they come with complications, including neurotoxicity, nephrotoxicity, and cardiotoxicity. Among these side effects, cardiotoxicity is the leading cause of morbidity and mortality, with anthracyclines being the primary culprit. Chemotherapy medications have various mechanisms that can lead to cardiac injury. Hence, numerous studies have been conducted to decrease the cardiotoxicity of these treatments. Combination therapy with beta-blockers, Angiotensin-converting enzyme inhibitors, and angiotensin receptor blockers have effectively reduced such outcomes. However, a definitive preventive strategy is yet to be established. Meanwhile, sodium-glucose co-transporter-2 (SGLT-2) inhibitors lower blood glucose levels in type 2 diabetes by reducing its re-absorption in the kidneys. They are thus considered potent drugs for glycemic control and reduction of cardiovascular risks. Recent studies have shown that SGLT-2 inhibitors are crucial in preventing chemotherapy-induced cardiotoxicity. They enhance heart cell viability, prevent degenerative changes, stimulate autophagy, and reduce cell death. This drug class also reduces inflammation by inhibiting reactive oxygen species and inflammatory cytokine production. Moreover, it can not only reverse the harmful effects of anticancer agents on the heart structure but also enhance the effectiveness of chemotherapy by minimizing potential consequences on the heart. In conclusion, SGLT-2 inhibitors hold promise as a therapeutic strategy for protecting cancer patients from chemotherapy-induced heart damage and improving cardiovascular outcomes.
Collapse
|
24
|
Kanagasabai T, Dunbar Z, Ochoa SG, Farris T, Dhandayuthapani S, Wijeratne EMK, Gunatilaka AAL, Shanker A. Bortezomib in Combination with Physachenolide C Reduces the Tumorigenic Properties of KRAS mut/P53 mut Lung Cancer Cells by Inhibiting c-FLIP. Cancers (Basel) 2024; 16:670. [PMID: 38339421 PMCID: PMC10854725 DOI: 10.3390/cancers16030670] [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: 11/22/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Defects in apoptosis regulation are one of the classical features of cancer cells, often associated with more aggressiveness and failure to therapeutic options. We investigated the combinatorial antitumor effects of a natural product, physachenolide C (PCC) and bortezomib, in KRASmut/P53mut lung cancer cells and xenograft mice models. METHODS The in vitro anticancer effects of the bortezomib and PCC combination were investigated using cell viability, migration, and invasion assays in 344SQ, H23, and H358 cell lines. Furthermore, the effects of combination treatment on the critical parameters of cellular metabolism, including extracellular acidification rate (ECAR) and mitochondrial oxidative phosphorylation based on the oxygen consumption rate of cancer cells were assessed using Seahorse assay. Finally, the antitumor effect of the bortezomib (1 mg/kg) and PCC (10 mg/kg) combination was evaluated using xenograft mice models. RESULTS Our data showed that the bortezomib-PCC combination was more effective in reducing the viability of lung cancer cells in comparison with the individual treatments. Similarly, the combination treatment showed a significant inhibition of cell migration and invasion of cancer cells. Additionally, the key anti-apoptotic protein c-FLIP was significantly inhibited along with a substantial reduction in the key parameters of cellular metabolism in cancer cells. Notably, the bortezomib or PCC inhibited the tumor growth compared to the control group, the tumor growth inhibition was much more effective when bortezomib was combined with PCC in tumor xenograft mice models. CONCLUSION These findings demonstrate that PCC sensitizes cancer cells to bortezomib, potentially improving the antitumor effects against KRASmut/P53mut lung cancer cells, with an enhanced efficacy of combination treatments without causing significant side effects.
Collapse
Affiliation(s)
- Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.K.); (T.F.)
| | - Zerick Dunbar
- Department of Microbiology, Immunology & Physiology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Salvador González Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Tonie Farris
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.K.); (T.F.)
| | | | - E. M. Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture, Life and Environmental Sciences, The University of Arizona, Tucson, AZ 85719, USA; (E.M.K.W.)
| | - A. A. Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture, Life and Environmental Sciences, The University of Arizona, Tucson, AZ 85719, USA; (E.M.K.W.)
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
25
|
Shekar N, Vuong P, Kaur P. Analysing potent biomarkers along phytochemicals for breast cancer therapy: an in silico approach. Breast Cancer Res Treat 2024; 203:29-47. [PMID: 37726449 PMCID: PMC10771382 DOI: 10.1007/s10549-023-07107-7] [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: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE This research focused on the identification of herbal compounds as potential anti-cancer drugs, especially for breast cancer, that involved the recognition of Notch downstream targets NOTCH proteins (1-4) specifically expressed in breast tumours as biomarkers for prognosis, along with P53 tumour antigens, that were used as comparisons to check the sensitivity of the herbal bio-compounds. METHODS After investigating phytochemical candidates, we employed an approach for computer-aided drug design and analysis to find strong breast cancer inhibitors. The present study utilized in silico analyses and protein docking techniques to characterize and rank selected bio-compounds for their efficiency in oncogenic inhibition for use in precise carcinomic cell growth control. RESULTS Several of the identified phytocompounds found in herbs followed Lipinski's Rule of Five and could be further investigated as potential medicinal molecules. Based on the Vina score obtained after the docking process, the active compound Epigallocatechin gallate in green tea with NOTCH (1-4) and P53 proteins showed promising results for future drug repurposing. The stiffness and binding stability of green tea pharmacological complexes were further elucidated by the molecular dynamic simulations carried out for the highest scoring phytochemical ligand complex. CONCLUSION The target-ligand complex of green tea active compound Epigallocatechin gallate with NOTCH (1-4) had the potential to become potent anti-breast cancer therapeutic candidates following further research involving wet-lab experiments.
Collapse
Affiliation(s)
- Nivruthi Shekar
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia
| | - Paton Vuong
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia
| | - Parwinder Kaur
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia.
| |
Collapse
|
26
|
Alavi SE, Alharthi S, Alavi SZ, Raza A, Ebrahimi Shahmabadi H. Bioresponsive drug delivery systems. Drug Discov Today 2024; 29:103849. [PMID: 38052319 DOI: 10.1016/j.drudis.2023.103849] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
In this review, we highlight the potential of stimuli-responsive drug delivery systems (DDSs) to revolutionize healthcare. Through examining pH, temperature, enzyme, and redox responsiveness, the presented case studies highlight the precision and enhanced therapeutic outcomes achievable with these innovative systems. Challenges, such as complex design and bio-based material optimization, underscore the complete journey from bench to bedside. Clinical strides in magnetically and temperature-responsive systems hint at a promising future for healthcare. However, overcoming issues of stability, durability, penetration depth, sensitivity, and active targeting is crucial. The future envisions theranostic systems, amalgamating targeted therapy and diagnosis, for personalized healthcare. Bio-based materials emerge as pivotal, offering a nuanced approach to complex diseases, such as cancer and diabetes, reshaping the healthcare landscape.
Collapse
Affiliation(s)
- Seyed Ebrahim Alavi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7718175911, Iran.
| | - Sitah Alharthi
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, Al-Dawadmi 11961, Saudi Arabia
| | - Seyed Zeinab Alavi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7718175911, Iran
| | - Aun Raza
- School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Hasan Ebrahimi Shahmabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7718175911, Iran.
| |
Collapse
|
27
|
Buaban K, Innets B, Petsri K, Sinsook S, Chanvorachote P, Chansriniyom C, Suwanborirux K, Yokoya M, Saito N, Chamni S. Semisynthesis of 5-O-ester derivatives of renieramycin T and their cytotoxicity against non-small-cell lung cancer cell lines. Sci Rep 2023; 13:21485. [PMID: 38057385 PMCID: PMC10700347 DOI: 10.1038/s41598-023-48526-2] [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: 03/19/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
The semisynthesis of 5-O-ester derivatives of renieramycin T was accomplished through the photoredox reaction of renieramycin M (1), a bistetrahydroisoquinolinequinone alkaloid isolated from the Thai blue sponge Xestospongia sp. This process led to the conversion of compound 1 to renieramycin T (2), which was subsequently subjected to Steglich esterification with appropriate acylating agents containing linear alkyl, N-tert-butoxycarbonyl-L-amino, and heterocyclic aromatic substituent. Notably, the one-pot transformation, combining the photoredox reaction and esterification led to the formation of 7-O-ester derivatives of renieramycin S due to hydrolysis. Subsequently, the in vitro cytotoxicity of the 17 semisynthesized derivatives against human non-small-cell lung cancer (NSCLC) cells in parallel with normal cell lines was evaluated. Among the tested compounds, 5-O-(3-propanoyl) ester of renieramycin T (3b) exhibited potent cytotoxic activity with half-maximal inhibitory concentration (IC50) values at 33.44 and 33.88 nM against H292 and H460 cell lines, respectively. These values were within the same range as compound 1 (IC50 = 34.43 and 35.63 nM) and displayed twofold higher cytotoxicity compared to compound 2 (IC50 = 72.85 and 83.95 nM). The steric characteristics and aromatic orientation of the 5-O-ester substituents played significant roles in their cytotoxicity. Notably, derivative 3b induced apoptosis with minimal necrosis, in contrast to the parental compound 1. Hence, the relationship between the structure and cytotoxicity of renieramycin-ecteinascidin hybrid alkaloids was investigated. This study emphasizes the potential of the series of 5-O-ester derivatives of renieramycin T as promising leads for the further development of potential anti-NSCLC agents.
Collapse
Affiliation(s)
- Koonchira Buaban
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Bhurichaya Innets
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Korrakod Petsri
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Suwimon Sinsook
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pithi Chanvorachote
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chaisak Chansriniyom
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Khanit Suwanborirux
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Masashi Yokoya
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Naoki Saito
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
28
|
Belov K, Brel V, Sobornova V, Fedorova I, Khodov I. Conformational Analysis of 1,5-Diaryl-3-Oxo-1,4-Pentadiene Derivatives: A Nuclear Overhauser Effect Spectroscopy Investigation. Int J Mol Sci 2023; 24:16707. [PMID: 38069031 PMCID: PMC10706324 DOI: 10.3390/ijms242316707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
1,5-Diaryl-3-Oxo-1,4-Pentadiene derivatives are intriguing organic compounds with a unique structure featuring a pentadiene core, aryl groups, and a ketone group. This study investigates the influence of fluorine atoms on the conformational features of these derivatives in deuterated chloroform (CDCl3) solution. Through nuclear magnetic resonance (NMR) spectroscopy and quantum chemical calculations, we discerned variations in interatomic distances and established predominant conformer proportions. The findings suggest that the non-fluorinated entity exhibits a uniform distribution across various conformer groups. The introduction of a fluorine atom induces substantial alterations, resulting in the predominance of a specific conformer group. This structural insight may hold the key to their diverse anticancer activities, previously reported in the literature.
Collapse
Affiliation(s)
- Konstantin Belov
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, 153045 Ivanovo, Russia; (K.B.); (V.S.); (I.F.)
| | - Valery Brel
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Valentina Sobornova
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, 153045 Ivanovo, Russia; (K.B.); (V.S.); (I.F.)
| | - Irina Fedorova
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, 153045 Ivanovo, Russia; (K.B.); (V.S.); (I.F.)
| | - Ilya Khodov
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, 153045 Ivanovo, Russia; (K.B.); (V.S.); (I.F.)
| |
Collapse
|
29
|
Azimi M, Yee SW, Riselli A, Silva DB, Giacomini CP, Giacomini KM, Brett CM. Characterization of P-glycoprotein orthologs from human, sheep, pig, dog, and cat. J Vet Pharmacol Ther 2023; 46:401-412. [PMID: 37198956 DOI: 10.1111/jvp.13386] [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: 12/20/2022] [Revised: 03/29/2023] [Accepted: 04/25/2023] [Indexed: 05/19/2023]
Abstract
The ATP-binding cassette transporter P-glycoprotein (P-gp) limits the oral bioavailability of many drugs. Although P-gp has been well studied in humans and mice, little is known about the substrate specificities of many of its species orthologs. To address this, we performed in vitro analysis of P-gp transporter function using HEK293 cells stably expressing human, ovine, porcine, canine, and feline P-gp. We also employed a human physiologically based pharmacokinetic (PBPK) model to assess variations in digoxin exposure resulting from altered P-gp function. Compared to human P-gp, sheep P-gp had significantly less digoxin efflux (2.3-fold ±0.04 vs. 1.8-fold ±0.03, p < .0001) and all species orthologs had significantly less quinidine efflux compared with human P-gp (p < .05). Human P-gp also had significantly greater efflux of talinolol compared to sheep and dog P-gp (1.9-fold ±0.04 vs. 1.6-fold ±0.06, p = .003 and 1.6-fold ±0.05, p = .0002, respectively). P-gp expression protected all lines against paclitaxel-induced toxicity, with sheep P-gp being significantly less protective. The inhibitor verapamil demonstrated dose-dependent inhibition of all P-gp orthologs. Finally, a PBPK model showed digoxin exposure was sensitive to altered P-gp activity. Overall, our study found that species differences in this major drug transporter exist and that the appropriate species ortholog of P-gp should be evaluated during veterinary drug development.
Collapse
Affiliation(s)
- Mina Azimi
- Apricity Therapeutics, Inc., San Francisco, California, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Sook Wah Yee
- Apricity Therapeutics, Inc., San Francisco, California, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Andrew Riselli
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Dina Buitrago Silva
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | | | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Claire M Brett
- Apricity Therapeutics, Inc., San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
30
|
Yehia Abdelzaher W, A Abdel-Gaber S, Atef Fawzy M, Hamid Sayed Abo Bakr Ali A, Ezzat Attya M, Geddawy A. Atorvastatin protects against cyclophosphamide-induced thyroid injury in rats via modulation of JNK/ ERK/ p38 MAPK signaling pathway. Int Immunopharmacol 2023; 124:111061. [PMID: 37844467 DOI: 10.1016/j.intimp.2023.111061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Cancer chemotherapy is associated with various tissue toxicities that limit its use. Cyclophosphamide (CYC) is one of the most commonly used antineoplastic and immunosuppressive agent. Thyroid dysfunction is a critical side effect of anticancer drugs. Atorvastatin (ATV) is antihyperlipedemic drug with different tissue protective activities. The aim of this study was to determine the potential protective effect of ATV against CYC-induced thyroid injury in rats. METHODS ATV was administered in the presence and absence of CYC. Thirty-two adult Wistar rats were randomly divided into four groups: control group, ATV group (20 mg/kg/day, p.o. for 14 day), CYC group (200 mg/kg, i.p. on day 9) and ATV/CYC group. Triiodothyronine (T3), thyroxine (T4), reduced glutathione (GSH), malondialdehyde (MDA), total nitrite/nitrate (NOx), p38 mitogen-activated protein kinase (P38MAPK), extracellular signal-regulated kinase (ERK) and c-Jun N-terminal Kinase (JNK) were measured. In addition, thyroid histopathology and caspase 3 immunohistochemistry were performed. RESULTS CYC significantly increased thyroid MDA, NOx, P38MAPK, ERK and JNK with decrease in GSH, T3 and T4 levels. Histopathological features of thyroid lesions and increased caspase 3 immune expression were appeared. ATV significantly normalized distributed oxidative, inflammatory and apoptotic indicators, resulting in an improvement of histopathological features and reduction of caspase 3 immunoexpression. CONCLUSION These findings suggest that ATV protects against CYC-induced thyroid injury by regulating the JNK/ERK/p38-MAPK signaling pathway.
Collapse
Affiliation(s)
| | - Seham A Abdel-Gaber
- Department of Pharmacology, Faculty of Medicine Minia University, Minia 61111, Egypt.
| | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | | | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia 61111, Egypt.
| | - Ayman Geddawy
- Department of Pharmacology, Faculty of Medicine Minia University, Minia 61111, Egypt; Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia.
| |
Collapse
|
31
|
Bérard C, Truillet C, Larrat B, Dhermain F, Estève MA, Correard F, Novell A. Anticancer drug delivery by focused ultrasound-mediated blood-brain/tumor barrier disruption for glioma therapy: From benchside to bedside. Pharmacol Ther 2023; 250:108518. [PMID: 37619931 DOI: 10.1016/j.pharmthera.2023.108518] [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: 07/17/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
The therapeutic management of gliomas remains particularly challenging. Brain tumors present multiple obstacles that make therapeutic innovation complex, mainly due to the presence of blood-tumor and blood-brain barriers (BTB and BBB, respectively) which prevent penetration of anticancer agents into the brain parenchyma. Focused ultrasound-mediated BBB disruption (FUS-BBBD) provides a physical method for non-invasive, local, and reversible BBB disruption. The safety of this technique has been demonstrated in small and large animal models. This approach promises to enhance drug delivery into the brain tumor and therefore to improve survival outcomes by repurposing existing drugs. Several clinical trials continue to be initiated in the last decade. In this review, we provide an overview of the rationale behind the use of FUS-BBBD in gliomas and summarize the preclinical studies investigating different approaches (free drugs, drug-loaded microbubbles and drug-loaded nanocarriers) in combination with this technology in in vivo glioma models. Furthermore, we discuss the current state of clinical trials and devices developed and review the challenges to overcome for clinical use of FUS-BBBD in glioma therapy.
Collapse
Affiliation(s)
- Charlotte Bérard
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91401 Orsay, France.
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Centre d'études de Saclay, 91191 Gif-sur-Yvette, France.
| | - Frédéric Dhermain
- Radiation Oncology Department, Gustave Roussy University Hospital, 94805 Villejuif, France.
| | - Marie-Anne Estève
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Florian Correard
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91401 Orsay, France.
| |
Collapse
|
32
|
Akash S, Bibi S, Biswas P, Mukerjee N, Khan DA, Hasan MN, Sultana NA, Hosen ME, Jardan YAB, Nafidi HA, Bourhia M. Revolutionizing anti-cancer drug discovery against breast cancer and lung cancer by modification of natural genistein: an advanced computational and drug design approach. Front Oncol 2023; 13:1228865. [PMID: 37817764 PMCID: PMC10561655 DOI: 10.3389/fonc.2023.1228865] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/15/2023] [Indexed: 10/12/2023] Open
Abstract
Breast and lung cancer are two of the most lethal forms of cancer, responsible for a disproportionately high number of deaths worldwide. Both doctors and cancer patients express alarm about the rising incidence of the disease globally. Although targeted treatment has achieved enormous advancements, it is not without its drawbacks. Numerous medicines and chemotherapeutic drugs have been authorized by the FDA; nevertheless, they can be quite costly and often fall short of completely curing the condition. Therefore, this investigation has been conducted to identify a potential medication against breast and lung cancer through structural modification of genistein. Genistein is the active compound in Glycyrrhiza glabra (licorice), and it exhibits solid anticancer efficiency against various cancers, including breast cancer, lung cancer, and brain cancer. Hence, the design of its analogs with the interchange of five functional groups-COOH, NH2 and OCH3, Benzene, and NH-CH2-CH2-OH-have been employed to enhance affinities compared to primary genistein. Additionally, advanced computational studies such as PASS prediction, molecular docking, ADMET, and molecular dynamics simulation were conducted. Firstly, the PASS prediction spectrum was analyzed, revealing that the designed genistein analogs exhibit improved antineoplastic activity. In the prediction data, breast and lung cancer were selected as primary targets. Subsequently, other computational investigations were gradually conducted. The mentioned compounds have shown acceptable results for in silico ADME, AMES toxicity, and hepatotoxicity estimations, which are fundamental for their oral medication. It is noteworthy that the initial binding affinity was only -8.7 kcal/mol against the breast cancer targeted protein (PDB ID: 3HB5). However, after the modification of the functional group, when calculating the binding affinities, it becomes apparent that the binding affinities increase gradually, reaching a maximum of -11.0 and -10.0 kcal/mol. Similarly, the initial binding affinity was only -8.0 kcal/mol against lung cancer (PDB ID: 2P85), but after the addition of binding affinity, it reached -9.5 kcal/mol. Finally, a molecular dynamics simulation was conducted to study the molecular models over 100 ns and examine the stability of the docked complexes. The results indicate that the selected complexes remain highly stable throughout the 100-ns molecular dynamics simulation runs, displaying strong correlations with the binding of targeted ligands within the active site of the selected protein. It is important to further investigate and proceed to clinical or wet lab experiments to determine the practical value of the proposed compounds.
Collapse
Affiliation(s)
- Shopnil Akash
- Faculty of Allied Health Science, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Nobendu Mukerjee
- Department of Microbiology, West Bengal State University, Kolkata, India
| | - Dhrubo Ahmed Khan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Nazneen Ahmeda Sultana
- Faculty of Allied Health Science, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Md. Eram Hosen
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Yousef A. Bin Jardan
- Department of Food Science, Faculty of Agricultural and Food Sciences, Laval University, Quebec City, QC, Canada
| | - Hiba-Allah Nafidi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, Ibn Zohr University, Laayoune, Morocco
| |
Collapse
|
33
|
Ouzounis S, Panagiotopoulos V, Bafiti V, Zoumpoulakis P, Cavouras D, Kalatzis I, Matsoukas MT, Katsila T. A Robust Machine Learning Framework Built Upon Molecular Representations Predicts CYP450 Inhibition: Toward Precision in Drug Repurposing. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023. [PMID: 37406257 PMCID: PMC10357106 DOI: 10.1089/omi.2023.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Human cytochrome P450 (CYP450) enzymes play a crucial role in drug metabolism and pharmacokinetics. CYP450 inhibition can lead to toxicity, in particular when drugs are co-administered with other drugs and xenobiotics or in the case of polypharmacy. Predicting CYP450 inhibition is also important for rational drug discovery and development, and precision in drug repurposing. In this overarching context, digital transformation of drug discovery and development, for example, using machine and deep learning approaches, offers prospects for prediction of CYP450 inhibition through computational models. We report here the development of a majority-voting machine learning framework to classify inhibitors and noninhibitors for seven major human liver CYP450 isoforms (CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). For the machine learning models reported herein, we employed interaction fingerprints that were derived from molecular docking simulations, thus adding an additional layer of information for protein-ligand interactions. The proposed machine learning framework is based on the structure of the binding site of isoforms to produce predictions beyond previously reported approaches. Also, we carried out a comparative analysis so as to identify which representation of test compounds (molecular descriptors, molecular fingerprints, or protein-ligand interaction fingerprints) affects the predictive performance of the models. This work underlines the ways in which the structure of the enzyme catalytic site influences machine learning predictions and the need for robust frameworks toward better-informed predictions.
Collapse
Affiliation(s)
- Sotiris Ouzounis
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- Department of Biomedical Engineering, University of West Attica, Egaleo, Greece
- Cloudpharm PC, Athens, Greece
| | - Vasilis Panagiotopoulos
- Department of Biomedical Engineering, University of West Attica, Egaleo, Greece
- Cloudpharm PC, Athens, Greece
| | - Vivi Bafiti
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Panagiotis Zoumpoulakis
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- Department of Food Science and Technology, University of West Attica, Egaleo, Greece
| | - Dionisis Cavouras
- Department of Biomedical Engineering, University of West Attica, Egaleo, Greece
| | - Ioannis Kalatzis
- Department of Biomedical Engineering, University of West Attica, Egaleo, Greece
| | - Minos-Timotheos Matsoukas
- Department of Biomedical Engineering, University of West Attica, Egaleo, Greece
- Cloudpharm PC, Athens, Greece
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| |
Collapse
|
34
|
Bou Malhab LJ, Bajbouj K, Shehab NG, Elayoty SM, Sinoj J, Adra S, Taneera J, Saleh MA, Abdel-Rahman WM, Semreen MH, Alzoubi KH, Bustanji Y, El-Huneidi W, Abu-Gharbieh E. Potential anticancer properties of calotropis procera: An investigation on breast and colon cancer cells. Heliyon 2023; 9:e16706. [PMID: 37332907 PMCID: PMC10272338 DOI: 10.1016/j.heliyon.2023.e16706] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Calotropis procera is a perennial flowering plant of the Apocynaceae family, traditionally used in medicine to treat various ailments. Recent investigations have revealed its potential therapeutic activities such as anti-inflammatory, gastroprotective, analgesic, anti-obesity, and anti-diabetic properties. RP-HPLC qualitatively and quantitatively evaluated the phenolic acids and flavonoids in the ethanolic extract at two different wavelengths, 280 and 330 nm. In addition, total phenolic and flavonoid contents were measured via spectrophotometric determination in addition to the antioxidant activity. The antiproliferative effects of C. procera were investigated on two cancer cell lines: human colon (HCT-116) and breast (MCF-7) cancer. Several methods were utilised to analyse the effectiveness of the plant extract on the cytotoxicity, apoptosis, cell cycle progression, genes involved in the cell cycle, and protein expression profiles of HCT-116 and MCF-7 cells. These included the MTT assay, Annexin V-FITC/PI, analysis of the cell cycle, and Western blot. Results indicated that ferulic and caffeic acids were the major compounds at λmax 280 nm (1.374% and 0.561%, respectively), while the major compounds at λmax 325 nm were kaempferol and luteolin (1.036% and 0.512%, respectively). The ethanolic extract had significantly higher antioxidant activity (80 ± 2.3%) compared to ascorbic acid (90 ± 3.1%). C. procera extract exhibited dose-dependent cell growth inhibition, with an estimated IC50 of 50 μg/mL for MCF-7 and 55 μg/mL for HCT-116 cells at 24 h. Annexin V-FITC/PI confirmed the induction of apoptosis. Remarkably, cell cycle arrest occurred at the sub-G1 phase in MCF-7 cells, while in HCT-116 cells, it was observed at the G2-M phase. The sub-G1 arrest was associated with dysregulation of Akt, p-AKT, mTOR, and p-mTOR proteins, as confirmed by the Western blot analysis, while downregulation of CDK1, cyclin B1, and survivin caused G2-M arrest.
Collapse
Affiliation(s)
- Lara J. Bou Malhab
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
| | - Khuloud Bajbouj
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Naglaa G. Shehab
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College, Dubai, 19099, United Arab Emirates
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza; 12613, Egypt
| | - Salma M. Elayoty
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College, Dubai, 19099, United Arab Emirates
| | - Jithna Sinoj
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
| | - Saryia Adra
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah; 27272, United Arab Emirates
| | - Jalal Taneera
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Mohamed A. Saleh
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah; 27272, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Wael M. Abdel-Rahman
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Karem H. Alzoubi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Departement of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
| | - Yasser Bustanji
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Waseem El-Huneidi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah; 27272, United Arab Emirates
| |
Collapse
|
35
|
Dorost P, García-Alvarez M, Martínez de Ilarduya A. Hydrophobic Modification of Poly(γ-glutamic acid) by Grafting 4-Phenyl-butyl Side Groups for the Encapsulation and Release of Doxorubicin. Pharmaceutics 2023; 15:pharmaceutics15051377. [PMID: 37242619 DOI: 10.3390/pharmaceutics15051377] [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: 02/28/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The delivery of drugs is a great challenge, since most of active pharmaceutical ingredients developed today are hydrophobic and poorly water soluble. From this perspective, drug encapsulation on biodegradable and biocompatible polymers can surpass this problem. Poly(γ-glutamic acid) (PGGA), a bioedible and biocompatible polymer has been chosen for this purpose. Carboxylic side groups of PGGA have been partially esterified with 4-phenyl-butyl bromide, producing a series of aliphatic-aromatic ester derivatives with different hydrophilic-lipophilic balances. Using nanoprecipitation or emulsion/evaporation methods, these copolymers were self-assembled in a water solution, forming nanoparticles with average diameters between 89 and 374 nm and zeta potential values between -13.1 and -49.5 mV. The hydrophobic core containing 4-phenyl-butyl side groups was used for the encapsulation of an anticancer drug, such as Doxorubicin (DOX). The highest encapsulation efficiency was reached for a copolymer derived from PGGA, with a 46 mol% degree of esterification. Drug release studies carried out for 5 days at different pHs (4.2 and 7.4) indicated that DOX was released faster at pH 4.2, revealing the potential of these nanoparticles as chemotherapy agents.
Collapse
Affiliation(s)
- Porochista Dorost
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, ETSEIB, Diagonal 647, 08028 Barcelona, Spain
| | - Montserrat García-Alvarez
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, ETSEIB, Diagonal 647, 08028 Barcelona, Spain
| | - Antxon Martínez de Ilarduya
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, ETSEIB, Diagonal 647, 08028 Barcelona, Spain
| |
Collapse
|
36
|
Miranda de Souza Duarte-Filho LA, Ortega de Oliveira PC, Yanaguibashi Leal CE, de Moraes MC, Picot L. Ligand fishing as a tool to screen natural products with anticancer potential. J Sep Sci 2023:e2200964. [PMID: 36808885 DOI: 10.1002/jssc.202200964] [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: 11/24/2022] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
Cancer is the second leading cause of death in the world and its incidence is expected to increase with the aging of the world's population and globalization of risk factors. Natural products and their derivatives have provided a significant number of approved anticancer drugs and the development of robust and selective screening assays for the identification of lead anticancer natural products are essential in the challenge of developing personalized targeted therapies tailored to the genetic and molecular characteristics of tumors. To this end, a ligand fishing assay is a remarkable tool to rapidly and rigorously screen complex matrices, such as plant extracts, for the isolation and identification of specific ligands that bind to relevant pharmacological targets. In this paper, we review the application of ligand fishing with cancer-related targets to screen natural product extracts for the isolation and identification of selective ligands. We provide critical analysis of the system configurations, targets, and key phytochemical classes related to the field of anticancer research. Based on the data collected, ligand fishing emerges as a robust and powerful screening system for the rapid discovery of new anticancer drugs from natural resources. It is currently an underexplored strategy according to its considerable potential.
Collapse
Affiliation(s)
| | | | - Cíntia Emi Yanaguibashi Leal
- Departamento de Ciências Farmacêuticas, Pós-Graduação em Biociências (PGB) Universidade Federal do Vale do São Francisco, Petrolina, Brazil
| | - Marcela Cristina de Moraes
- Departamento de Química Orgânica, Laboratório BIOCROM, Instituto de Química, Universidade Federal Fluminense, Niterói, Brazil
| | - Laurent Picot
- UMR CNRS 7266 LIENSs, Département de Biotechnologie, La Rochelle Université, La Rochelle, France
| |
Collapse
|
37
|
K Johnson K, Kopecky C, Koshy P, Liu Y, Devadason M, Holst J, A Kilian K, C Sorrell C. Theranostic Activity of Ceria-Based Nanoparticles toward Parental and Metastatic Melanoma: 2D vs 3D Models. ACS Biomater Sci Eng 2023; 9:1053-1065. [PMID: 36726306 DOI: 10.1021/acsbiomaterials.2c01258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The time interval between the diagnosis of tumor in a patient and the initiation of treatment plays a key role in determining the survival rates. Consequently, theranostics, which is a combination of diagnosis and treatment, can be expected to improve survival rates. Early detection and immediate treatment initiation are particularly important in the management of melanoma, where survival rates decrease considerably after metastasis. The present work reports for the first time the application of fluorescein isothiocyanate (FITC)-tagged epidermal growth factor receptor (EGFR)-functionalized ceria nanoparticles, which exhibit intrinsic reactive oxygen species (ROS)-mediated anticancer effects, for the EGFR-targeted diagnosis and treatment of melanoma. The theranostic activity was demonstrated using two-dimensional (2D) and three-dimensional (3D) models of parental and metastatic melanoma. Confocal imaging studies confirm the diagnostic activity of the system. The therapeutic efficiency was evaluated using cell viability studies and ROS measurements. The ROS elevation levels are compared across the 2D and 3D models. Significant enhancement in the generation of cellular ROS and absence in mitochondrial ROS are observed in the 2D models. In contrast, significant elevations in both ROS types are observed for the 3D models, which are significantly higher for the metastatic spheroids than the parental spheroids, thus indicating the suitability of this nanoformulation for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Kochurani K Johnson
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Chantal Kopecky
- Australian Centre for NanoMedicine, School of Chemistry, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Pramod Koshy
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Yiling Liu
- Australian Centre for NanoMedicine, School of Chemistry, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Michelle Devadason
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jeff Holst
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Kristopher A Kilian
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia.,Australian Centre for NanoMedicine, School of Chemistry, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Charles C Sorrell
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
38
|
The Role of Silver Nanoparticles in the Diagnosis and Treatment of Cancer: Are There Any Perspectives for the Future? Life (Basel) 2023; 13:life13020466. [PMID: 36836823 PMCID: PMC9965924 DOI: 10.3390/life13020466] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Cancer is a fatal disease with a complex pathophysiology. Lack of specificity and cytotoxicity, as well as the multidrug resistance of traditional cancer chemotherapy, are the most common limitations that often cause treatment failure. Thus, in recent years, significant efforts have concentrated on the development of a modernistic field called nano-oncology, which provides the possibility of using nanoparticles (NPs) with the aim to detect, target, and treat cancer diseases. In comparison with conventional anticancer strategies, NPs provide a targeted approach, preventing undesirable side effects. What is more, nanoparticle-based drug delivery systems have shown good pharmacokinetics and precise targeting, as well as reduced multidrug resistance. It has been documented that, in cancer cells, NPs promote reactive oxygen species (ROS) production, induce cell cycle arrest and apoptosis, activate ER (endoplasmic reticulum) stress, modulate various signaling pathways, etc. Furthermore, their ability to inhibit tumor growth in vivo has also been documented. In this paper, we have reviewed the role of silver NPs (AgNPs) in cancer nanomedicine, discussing numerous mechanisms by which they render anticancer properties under both in vitro and in vivo conditions, as well as their potential in the diagnosis of cancer.
Collapse
|
39
|
Muthukumaran MK, Govindaraj M, Raja BK, J AS. In situ synthesis of polythiophene encapsulated 2D hexagonal boron nitride nanocomposite based electrochemical transducer for detection of 5-fluorouracil with high selectivity. RSC Adv 2023; 13:2780-2794. [PMID: 36756436 PMCID: PMC9850362 DOI: 10.1039/d2ra07147a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
It is difficult for the scientific community to develop a nonenzymatic sensing platform for extremely sensitive and selective detection of specific biomolecules, antibiotics, food adulterants, heavy metals, etc. One of the most significant chemotherapy drugs, 5-fluorouracil (5-Fu), which is used to treat solid malignancies, has a fluorine atom in the fifth position of the uracil molecule. Recognizing the secure and effective dosing of drugs for chemotherapy continues to be a critical concern in cancer disease management. The maintenance of the optimal 5-Fu concentration is dependent on the presence of 5-Fu in biofluids. Herein we reported a conducting polymer encapsulated 2D material, PTh/h-BN for the efficient electrochemical detection of anticancer drug 5-Fu. Furthermore, the synthesized PTh/h-BN nanocomposite was confirmed by the High-Resolution Transmission Electron Microscope (HR-TEM), High-Resolution Scanning Electron Microscope (HR-SEM), X-ray diffraction (XRD), and Fourier-Transform Infrared Spectroscopy (FT-IR). The electrical resistance of PTh/h-BN modified GCE and its sensing performance towards 5-Fu were tested using Electrochemical Impedance Spectroscopy (EIS) and Cyclic Voltammetry (CV) studies respectively. The analytical performance of our proposed catalyst was tested using Differential Pulse Voltammetry (DPV), and the amperometry (i-t curve) method. From the results, our proposed PTh/h-BN nanocomposite-modified GCE shows enhanced sensing performance due to higher redox peak currents, large active surface area, and high electrical conductivity. Moreover, the nanohybrid shows enhanced sensing performances with quick response time, wide linear range, the lowest limit of detection, high sensitivity, and high selectivity in the presence of various interferents. Finally, the practical applicability of the proposed sensor was tested with real-world samples with very good recovery percentages.
Collapse
Affiliation(s)
- Magesh Kumar Muthukumaran
- Department of Chemistry, SRM Institute of Science and Technology Kattankulathur 603203 Tamil Nadu India
| | - Muthukumar Govindaraj
- Department of Chemistry, SRM Institute of Science and Technology Kattankulathur 603203 Tamil Nadu India
| | - Bharathi Kannan Raja
- Department of Chemistry, SRM Institute of Science and Technology Kattankulathur 603203 Tamil Nadu India
| | - Arockia Selvi J
- Department of Chemistry, SRM Institute of Science and Technology Kattankulathur 603203 Tamil Nadu India
| |
Collapse
|
40
|
Ibrahim D, Abozied N, Abdel Maboud S, Alzamami A, Alturki NA, Jaremko M, Alanazi MK, Alhuthali HM, Seddek A. Therapeutic potential of bone marrow mesenchymal stem cells in cyclophosphamide-induced infertility. Front Pharmacol 2023; 14:1122175. [PMID: 37033609 PMCID: PMC10073512 DOI: 10.3389/fphar.2023.1122175] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/26/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer is a deadly disease characterized by abnormal cell proliferation. Chemotherapy is one technique of cancer treatment. Cyclophosphamide (CYP) is the most powerful chemotherapy medication, yet it has serious adverse effects. It is an antimitotic medicine that regulates cell proliferation and primarily targets quickly dividing cells, and it has been related to varying levels of infertility in humans. In the current study, we assessed the biochemical, histological, and microscopic evaluations of testicular damage following cyclophosphamide administration. Further, we have explored the potential protective impact of mesenchymal stem cell (MSCs) transplantation. The biochemical results revealed that administration of cyclophosphamide increased serum concentrations of follicle-stimulating hormone (FSH) and luteinizing hormone (LH), while it decreased serum concentrations of free testosterone hormone (TH), testicular follicle-stimulating hormone, luteinizing hormone, and free testosterone hormone concentrations, testicular total antioxidant capacity (TAC), and testicular activity of superoxide dismutase (SOD) enzyme. The histology and sperm examinations revealed that cyclophosphamide induced destruction to the architectures of several tissues in the testes, which drastically reduced the Johnsen score as well as the spermatogenesis process. Surprisingly, transplantation of mesenchymal stem cell after cyclophosphamide administration altered the deterioration effect of cyclophosphamide injury on the testicular tissues, as demonstrated by biochemical and histological analysis. Our results indicated alleviation of serum and testicular sex hormones, as well as testicular oxidative stress markers (total antioxidant capacity and superoxide dismutase activity), and nearly restored the normal appearance of the testicular tissues, Johnsen score, and spermatogenesis process. In conclusion, our work emphasizes the protective pharmacological use of mesenchymal stem cell to mitigate the effects of cyclophosphamide on testicular tissues that impair the spermatogenesis process following chemotherapy. These findings indicate that transferring mesenchymal stem cell to chemotherapy patients could significantly improve spermatogenesis.
Collapse
Affiliation(s)
- Dalia Ibrahim
- The Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- *Correspondence: Dalia Ibrahim,
| | - Nadia Abozied
- The Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Samar Abdel Maboud
- The Department of Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah, Saudi Arabia
| | - Norah A. Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative and Red Sea Research Center, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Maram Khalil Alanazi
- Pharm.D, Scientific Office and Regulatory Affair Department, Dallah Pharma Company, Riyadh, Saudi Arabia
| | - Hayaa M. Alhuthali
- Department of Clinical laboratory sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Asmaa Seddek
- The Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
41
|
Bhat AA, Gupta G, Alharbi KS, Afzal O, Altamimi ASA, Almalki WH, Kazmi I, Al-Abbasi FA, Alzarea SI, Chellappan DK, Singh SK, MacLoughlin R, Oliver BG, Dua K. Polysaccharide-Based Nanomedicines Targeting Lung Cancer. Pharmaceutics 2022; 14:pharmaceutics14122788. [PMID: 36559281 PMCID: PMC9782996 DOI: 10.3390/pharmaceutics14122788] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
A primary illness that accounts for a significant portion of fatalities worldwide is cancer. Among the main malignancies, lung cancer is recognised as the most chronic kind of cancer around the globe. Radiation treatment, surgery, and chemotherapy are some medical procedures used in the traditional care of lung cancer. However, these methods lack selectivity and damage nearby healthy cells. Several polysaccharide-based nanomaterials have been created to transport chemotherapeutics to reduce harmful and adverse side effects and improve response during anti-tumour reactions. To address these drawbacks, a class of naturally occurring polymers called polysaccharides have special physical, chemical, and biological characteristics. They can interact with the immune system to induce a better immunological response. Furthermore, because of the flexibility of their structures, it is possible to create multifunctional nanocomposites with excellent stability and bioavailability for the delivery of medicines to tumour tissues. This study seeks to present new views on the use of polysaccharide-based chemotherapeutics and to highlight current developments in polysaccharide-based nanomedicines for lung cancer.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School and of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Gaurav Gupta
- School and of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
- Correspondence:
| | - Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen, IDA Business Park, Dangan, H91 HE94 Galway, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology, Sydney, NSW 2007, Australia
- Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW 2000, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
42
|
Synthesis and Preclinical Evaluation of Small-Molecule Prostate-Specific Membrane Antigen-Targeted Abiraterone Conjugate. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248795. [PMID: 36557929 PMCID: PMC9783881 DOI: 10.3390/molecules27248795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Prostate cancer is the second most common type of cancer among men. The main method of its treatment is androgen deprivation therapy, which has a wide range of side effects. One of the solutions to this challenge is the targeted delivery of drugs to prostate cancer cells. In this study, we performed the synthesis of a novel small-molecule PSMA-targeted conjugate based on abiraterone. Cytotoxicity, the induction of intracellular reactive oxygen species, and P450-cytochrome species inhibition were investigated for this conjugate PSMA-abiraterone. The conjugate demonstrated a preferential effect on prostate tumor cells, remaining inactive at up to 100 µM in human fibroblast cells. In addition, it revealed preferential efficacy, specifically on PSMA-expressing lines with a 65% tumor growth inhibition level on 22Rv1 (PSMA+) xenografts after 14-fold oral administration of PSMA-Abi at a single dose of 500 mg/kg (7.0 g/kg total dose) was observed. This compound showed significantly reduced acute toxicity with comparable efficacy compared to AbiAc.
Collapse
|
43
|
Shahriar SMS, Andrabi SM, Islam F, An JM, Schindler SJ, Matis MP, Lee DY, Lee YK. Next-Generation 3D Scaffolds for Nano-Based Chemotherapeutics Delivery and Cancer Treatment. Pharmaceutics 2022; 14:2712. [PMID: 36559206 PMCID: PMC9784306 DOI: 10.3390/pharmaceutics14122712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is the leading cause of death after cardiovascular disease. Despite significant advances in cancer research over the past few decades, it is almost impossible to cure end-stage cancer patients and bring them to remission. Adverse effects of chemotherapy are mainly caused by the accumulation of chemotherapeutic agents in normal tissues, and drug resistance hinders the potential therapeutic effects and curing of this disease. New drug formulations need to be developed to overcome these problems and increase the therapeutic index of chemotherapeutics. As a chemotherapeutic delivery platform, three-dimensional (3D) scaffolds are an up-and-coming option because they can respond to biological factors, modify their properties accordingly, and promote site-specific chemotherapeutic deliveries in a sustainable and controlled release manner. This review paper focuses on the features and applications of the variety of 3D scaffold-based nano-delivery systems that could be used to improve local cancer therapy by selectively delivering chemotherapeutics to the target sites in future.
Collapse
Affiliation(s)
- S. M. Shatil Shahriar
- Eppley Institute for Research in Cancer and Allied Diseases, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Surgery—Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Syed Muntazir Andrabi
- Department of Surgery—Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Farhana Islam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | | | - Mitchell P. Matis
- Kansas City Internal Medicine Residency Program, HCA Healthcare, Overland Park, KS 66215, USA
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul 04763, Republic of Korea
| | - Yong-kyu Lee
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
| |
Collapse
|
44
|
Jampilek J, Kralova K. Insights into Lipid-Based Delivery Nanosystems of Protein-Tyrosine Kinase Inhibitors for Cancer Therapy. Pharmaceutics 2022; 14:2706. [PMID: 36559200 PMCID: PMC9783038 DOI: 10.3390/pharmaceutics14122706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022] Open
Abstract
According to the WHO, cancer caused almost 10 million deaths worldwide in 2020, i.e., almost one in six deaths. Among the most common are breast, lung, colon and rectal and prostate cancers. Although the diagnosis is more perfect and spectrum of available drugs is large, there is a clear trend of an increase in cancer that ends fatally. A major advance in treatment was the introduction of gentler antineoplastics for targeted therapy-tyrosine kinase inhibitors (TKIs). Although they have undoubtedly revolutionized oncology and hematology, they have significant side effects and limited efficacy. In addition to the design of new TKIs with improved pharmacokinetic and safety profiles, and being more resistant to the development of drug resistance, high expectations are placed on the reformulation of TKIs into various drug delivery lipid-based nanosystems. This review provides an insight into the history of chemotherapy, a brief overview of the development of TKIs for the treatment of cancer and their mechanism of action and summarizes the results of the applications of self-nanoemulsifying drug delivery systems, nanoemulsions, liposomes, solid lipid nanoparticles, lipid-polymer hybrid nanoparticles and nanostructured lipid carriers used as drug delivery systems of TKIs obtained in vitro and in vivo.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| | - Katarina Kralova
- Institute of Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
45
|
Lian MQ, Chng WH, Liang J, Yeo HQ, Lee CK, Belaid M, Tollemeto M, Wacker MG, Czarny B, Pastorin G. Plant-derived extracellular vesicles: Recent advancements and current challenges on their use for biomedical applications. J Extracell Vesicles 2022; 11:e12283. [PMID: 36519808 PMCID: PMC9753580 DOI: 10.1002/jev2.12283] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EVs) represent a diverse class of lipid bilayer membrane vesicles released by both animal and plant cells. These ubiquitous vesicles are involved in intercellular communication and transport of various biological cargos, including proteins, lipids, and nucleic acids. In recent years, interest in plant-derived EVs has increased tremendously, as they serve as a scalable and sustainable alternative to EVs derived from mammalian sources. In vitro and in vivo findings have demonstrated that these plant-derived vesicles (PDVs) possess intrinsic therapeutic activities that can potentially treat diseases and improve human health. In addition, PDVs can also act as efficient and biocompatible drug carriers. While preclinical studies have shown promising results, there are still several challenges and knowledge gaps that have to be addressed for the successful translation of PDVs into clinical applications, especially in view of the lack of standardised protocols for material handling and PDV isolation from various plant sources. This review provides the readers with a quick overview of the current understanding and research on PDVs, critically analysing the current challenges and highlighting the immense potential of PDVs as a novel class of therapeutics to treat human diseases. It is expected that this work will guide scientists to address the knowledge gaps currently associated with PDVs and promote new advances in plant-based therapeutic solutions.
Collapse
Affiliation(s)
| | - Wei Heng Chng
- Department of PharmacyNational University of SingaporeSingaporeSingapore
- Integrative Sciences and Engineering Programme, NUS Graduate SchoolNational University of SingaporeSingaporeSingapore
| | - Jeremy Liang
- Department of ChemistryNational University of SingaporeSingaporeSingapore
| | - Hui Qing Yeo
- Department of PharmacyNational University of SingaporeSingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Choon Keong Lee
- Department of PharmacyNational University of SingaporeSingaporeSingapore
| | - Mona Belaid
- Institute of Pharmaceutical ScienceKing's College LondonLondonUnited Kingdom
| | - Matteo Tollemeto
- Department of Health TechnologyTechnical University of DenmarkKongens LyngbyDenmark
| | | | - Bertrand Czarny
- School of Materials Science & EngineeringNanyang Technological UniversitySingaporeSingapore
| | - Giorgia Pastorin
- Department of PharmacyNational University of SingaporeSingaporeSingapore
| |
Collapse
|
46
|
Saghafi A, Ghorbani M, Pakseresht M, Shams A. Synthesis and development of novel magnetic polymeric sorbent to simultaneous extraction of three anti-cancers using dispersive micro solid phase extraction procedure in biological and water samples. Microchem J 2022. [DOI: 10.1016/j.microc.2022.108047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
47
|
Liu Y, Ahmed Smait D, Yaseen Naser A, M. A. Altalbawy F, Bahri H, Abdul Kadhim Ruhaima A, Zayad Fathallah T, Hadrawi SK, Alsaddon RE, Alshetaili A, Alsubaiyel AM. Optimization and validation of drug solubility by development of advanced artificial intelligence models. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.121113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
48
|
Yu B, Wang X, Ding L, Han M, Guo Y. Hydrophilic Natural Polylysine as Drug Nanocarrier for Preparation of Helical Delivery System. Pharmaceutics 2022; 14:pharmaceutics14112512. [PMID: 36432704 PMCID: PMC9696163 DOI: 10.3390/pharmaceutics14112512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Polypeptide materials have clear secondary structure and biodegradability, which can be further modified and functionalized, so that they can be employed as therapeutic agents in clinical applications. PEGylation of polylysine (PEG-PLL) is a kind of safe and effective nanocarrier that is utilized for gene and drug delivery. However, PEG-PLL needs to be produced through chemical synthesis, which is expensive and difficult to obtain. We hope to simplify the nanocarrier and use hydrophilic natural polylysine (PLL) to develop a high-efficacy delivery system. To evaluate the possibility of PLL as nanocarriers, methotrexate (MTX) is selected as a model drug and PEG-PLL is utilized as control nanocarriers. The experimental results showed that PLL is an ideal polypeptide to prepare MTX-loaded PLL nanoparticles (PLL/MTX NPs). Compared with PEG-PLL as nanocarriers, PLL/MTX NPs showed higher drug-loading content (58.9%) and smaller particle sizes (113.7 nm). Moreover, the shape of PLL/MTX NPs was a unique helical nanorod. The PLL/MTX NPs had good storage stability, media stability, and sustained release effect. Animal research demonstrated that PLL/MTX NPs could improve the anti-tumor activity of MTX, the antitumor efficacy is enhanced 1.9-fold and 1.2-fold compared with MTX injection and PEG-PLL/MTX NPs, respectively. To sum up, natural polymer PLL is an ideal nano drug delivery carrier which has potential clinical applications.
Collapse
Affiliation(s)
- Bo Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Lijuan Ding
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Correspondence:
| |
Collapse
|
49
|
Shin HC, Kim Y, Choi J, Kang HB, Han SY, Park K, Hwang HJ. Regioselective Synthesis of 6- O-Acetyl Dieckol and Its Selective Cytotoxicity against Non-Small-Cell Lung Cancer Cells. Mar Drugs 2022; 20:683. [PMID: 36355006 PMCID: PMC9695823 DOI: 10.3390/md20110683] [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: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
Dieckol, a phlorotannin from Ecklonia cava, has shown potential for use as an anticancer agent that selectively kills cancer cells. However, it is necessary to amplify its potency without damaging its inherent safety in order to develop it as a competitive chemotherapeutic. Here, we explored the controlled O-acylations of dieckol. Acyl groups could be consistently introduced to the 6-O position of dieckol with a high regioselectivity, which was confirmed by NOESY, HMBC and HSQC spectroscopies. In cytotoxicity studies on the newly synthesized 6-O-acetyl, 6-O-benzoyl dieckols and previously synthesized 6-O-alkyl dieckols against A549 vs. normal cells, all of the derivatives showed low cytotoxicity in normal cells with an IC50 of 481-719 μM, and highly structure-dependent cytotoxicity in A549 cells with an IC50 of 7.02 (acetyl)-842.26 (benzyl) μM. The selectivity index also showed a large structure dependency in the range of 0.67 (benzyl)-68.58 (acetyl). An analysis of the structure-activity relationship indicated that the activity was dramatically reduced in the presence of a benzene ring and was highly increased in the presence of small polar substituents. Conclusions: Controlled mono-O-modifications of dieckol could be a powerful tool to enhance the anticancer activity of dieckol, thus contributing to the development strategy for dieckol-based chemotherapeutics.
Collapse
Affiliation(s)
- Hyeon-Cheol Shin
- Center for Molecular Intelligence, The State University of New York, Korea, 119 Songdomunhwa-ro, Yeonsu-gu, Incheon 21985, Korea
| | - Yongkyun Kim
- School of Chemical Engineering and Material Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 03722, Korea
| | - Jaeyeong Choi
- School of Chemical Engineering and Material Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 03722, Korea
| | - Hyun Bae Kang
- Healinnols Inc., 1662 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea
| | - Seung-Yun Han
- College of Medicine, Konyang University, 158 Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
| | - Kwangyong Park
- School of Chemical Engineering and Material Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 03722, Korea
| | - Hye Jeong Hwang
- Center for Molecular Intelligence, The State University of New York, Korea, 119 Songdomunhwa-ro, Yeonsu-gu, Incheon 21985, Korea
| |
Collapse
|
50
|
Bioanalysis by LC-MS/MS and preclinical pharmacokinetic interaction study of ribociclib and oleanolic acid. Bioanalysis 2022; 14:1051-1065. [PMID: 36148926 DOI: 10.4155/bio-2022-0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Ribociclib (RIBO), approved in 2017 for HR-positive and HER-2-negative metastatic breast cancer treatment is reported to have the potential to induce hepatobiliary toxicity in patients. Oleanolic acid (OLA) has hepatoprotective potential that can be beneficial if coadministered with RIBO. Methodology & results: The primary scope of this study was to develop quantitative bioanalytical methods for RIBO and OLA. Two methods (for +ve electrospray ionization [ESI] and -ve ESI) were developed and validated according to USFDA bioanalytical guidelines. Discussion/conclusion: A single and simple sample preparation method was developed with >75% recovery. The accuracy and precision for RIBO and OLA were within acceptable limits over the calibration range of 5-500 ng/ml. This work reports, for the first time, the drug-drug interaction potential between RIBO and OLA.
Collapse
|