201
|
Ruhul-Amin M, Rahman MA, Khatun N, Hasan I, Kabir SR, Asaduzzaman A. Bioactivity of biogenic silver/silver chloride nanoparticles from Maranta arundinacea rhizome extract: Antibacterial and antioxidant properties with anticancer potential against Ehrlich ascites carcinoma and human breast cancer cell lines. Heliyon 2024; 10:e39493. [PMID: 39502215 PMCID: PMC11535985 DOI: 10.1016/j.heliyon.2024.e39493] [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: 05/03/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction This study explores the synthesis and characterization of silver/silver chloride nanoparticles (Ag/AgCl-NPs) using Maranta arundinacea rhizome extract and evaluates their bioactivities, including antibacterial, antioxidant, and anticancer potentials. Methods The synthesis of Ag/AgCl-NPs was initially confirmed by a color change and a sharp peak at 463 nm in UV-visible spectroscopy. Further characterization was conducted using scanning electron microscopy (SEM), X-ray powder diffraction (XRD), and fourier transform infrared spectroscopy (FTIR). Antibacterial properties were checked against four pathogenic bacteria (Shigella boydii, Escherichia coli, Shigella dysenteriae, and Staphylococcus aureus), and antioxidant activities were assessed using DPPH (2,2-diphenyl-1-picrylhydrazyl) and ABTS (2,2-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid) assay. In addition, the anticancer potential was evaluated in vitro using MTT (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) colorimetric assay and in vivo using the mouse models. Finally, toxicity was determined by employing the brine shrimp nauplii lethality assay. Results Ag/AgCl-NPs most effectively inhibited the growth of Staphylococcus aureus, showing maximum zone of inhibition and 7 μg/mL of minimum inhibitory concentration (MIC), and prevented the biofilm formation by Escherichia coli at 40 μg/mL. They displayed antioxidant activities against DPPH and ABTS with IC50 values of 90.65 and 24.34 μg/mL, respectively. In vitro, they inhibited 61.96 % EAC and 49.63 % MCF-7 cells growth at 32 and 128 μg/mL, respectively. Subsequently, inhibition rates of EAC cells growth in mice were measured as 38.30 %, 57.38 %, and 31.81 % after employing 2.5, 5, and 10 mg/kg/day of Ag/AgCl-NPs, respectively. Moreover, Ag/AgCl-NPs treated mice were found to carry more apoptotic EAC cells with distorted morphology. Treated mice showed decreased tumor weight, increased mean survival time, and a lifespan increase of up to 30 %, with improved hematological parameters. Later, Ag/AgCl-NPs exhibited moderate toxicity with an LC50 value of 208.41 μg/mL in brine shrimp nauplii lethality assay. Conclusion The promising antibacterial, antioxidant, and anticancer activities along with mild toxicity suggest the potential biomedical uses of Maranta arundinacea rhizome extract-mediated Ag/AgCl-NPs.
Collapse
Affiliation(s)
- Md. Ruhul-Amin
- Department of Biochemistry & Molecular Biology, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry and Molecular Biology, Trust University, Barisal, 8200, Bangladesh
| | - Md. Abdur Rahman
- Department of Biochemistry & Molecular Biology, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Nisa Khatun
- Department of Biochemistry & Molecular Biology, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Imtiaj Hasan
- Department of Biochemistry & Molecular Biology, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Syed Rashel Kabir
- Department of Biochemistry & Molecular Biology, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - A.K.M. Asaduzzaman
- Department of Biochemistry & Molecular Biology, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| |
Collapse
|
202
|
Kot Y, Klochkov V, Prokopiuk V, Sedyh O, Tryfonyuk L, Grygorova G, Karpenko N, Tomchuk O, Kot K, Onishchenko A, Yefimova S, Tkachenko A. GdVO 4:Eu 3+ and LaVO 4:Eu 3+ Nanoparticles Exacerbate Oxidative Stress in L929 Cells: Potential Implications for Cancer Therapy. Int J Mol Sci 2024; 25:11687. [PMID: 39519237 PMCID: PMC11546343 DOI: 10.3390/ijms252111687] [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: 09/25/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
The therapeutic potential of redox-active nanoscale materials as antioxidant- or reactive oxygen species (ROS)-inducing agents was intensely studied. Herein, we demonstrate that the synthesized and characterized GdVO4:Eu3+ and LaVO4:Eu3+ nanoparticles, which have been already shown to have redox-active, anti-inflammatory, antibacterial, and wound healing properties, both in vitro and in vivo, worsen oxidative stress of L929 cells triggered by hydrogen peroxide or tert-butyl hydroperoxide (tBuOOH) at the concentrations that are safe for intact L929 cells. This effect was observed upon internalization of the investigated nanosized materials and is associated with the cleavage of caspase-3 and caspase-9 without recruitment of caspase-8. Such changes in the caspase cascade indicate activation of the intrinsic caspase-9-dependent mitochondrial but not the extrinsic death, receptor-mediated, and caspase-8-dependent apoptotic pathway. The GdVO4:Eu3+ and LaVO4:Eu3+ nanoparticle-induced apoptosis of oxidatively compromised L929 cells is mediated by ROS overgeneration, Ca2+ overload, endoplasmic reticulum stress-associated JNK (c-Jun N-terminal kinase), and DNA damage-inducible transcript 3 (DDIT3). Our findings demonstrate that GdVO4:Eu3+ and LaVO4:Eu3+ nanoparticles aggravate the oxidative stress-induced damage to L929 cells, indicating that they might potentially be applied as anti-cancer agents.
Collapse
Affiliation(s)
- Yuriy Kot
- Department of Biochemistry, V.N. Karazin Kharkiv National, 4 Svobody Sq, 61022 Kharkiv, Ukraine; (Y.K.); (K.K.)
| | - Vladimir Klochkov
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Volodymyr Prokopiuk
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavskaya Str., 61015 Kharkiv, Ukraine; (V.P.); (A.O.)
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, 4 Nauky Ave, 61022 Kharkiv, Ukraine
| | - Olha Sedyh
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Liliya Tryfonyuk
- Institute of Health, National University of Water and Environmental Engineering, 11 Soborna Str., 33028 Rivne, Ukraine;
| | - Ganna Grygorova
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Nina Karpenko
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Oleksandr Tomchuk
- ISIS Neutron and Muon Source, Rutherford Appleton Laboratory, Harwell Oxford, Didcot OX11 0QX, UK;
| | - Kateryna Kot
- Department of Biochemistry, V.N. Karazin Kharkiv National, 4 Svobody Sq, 61022 Kharkiv, Ukraine; (Y.K.); (K.K.)
| | - Anatolii Onishchenko
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavskaya Str., 61015 Kharkiv, Ukraine; (V.P.); (A.O.)
| | - Svetlana Yefimova
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky Ave, 61072 Kharkiv, Ukraine; (V.K.); (O.S.); (G.G.); (N.K.)
| | - Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 25250 Vestec, Czech Republic
| |
Collapse
|
203
|
Chatterjee A, Khan R, Mukherjee T, Sahoo PP, Tiwari LN, Singh BN, Kumari R, Kumari A, Rai A, Ray S. Harnessing bacterial metabolites for enhanced cancer chemotherapy: unveiling unique therapeutic potentials. Arch Microbiol 2024; 206:449. [PMID: 39472338 DOI: 10.1007/s00203-024-04179-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: 08/15/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
Cancer poses a serious threat to health globally, with millions diagnosed every year. According to Global Cancer Statistics 2024, about 20 million new cases were reported in 2022, and 9.7 million people worldwide died of this condition. Advanced therapies include combination of one or more treatment procedures, depending on the type, stage, and particular genetic constitution of the cancer, which may include surgery, radiotherapy, chemotherapy, immunotherapy, hormone therapy, targeted therapy, and stem cell transplant. Also, awareness about lifestyle changes, preventive measures and screening at early stages has reduced the incidence of the disease; still, there is a major failure in controlling the incidence of cancer because of its complex and multifaceted nature. With increasing interest in bacterial metabolites as possible novel and effective treatment options in cancer therapy, their main benefits include not only direct anticancer effects but also the modulation of the immune system and potential for targeted and combination therapies. They can therefore be used in combination with chemotherapy, radiotherapy, or immunotherapy to improve outcomes or reduce side effects. Furthermore, nanoparticle-based delivery systems have the potential to enhance the potency and safety of anticancer drugs by providing improved stability, targeted release, and controlled delivery.
Collapse
Affiliation(s)
- Aroni Chatterjee
- Department of Biotechnology, School of Biotechnology and Biosciences, Brainware University, Barasat, Kolkata, 700125, West Bengal, India
| | - Rajni Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, 844102, Bihar, India
| | - Triparna Mukherjee
- Department of Biotechnology, School of Biotechnology and Biosciences, Brainware University, Barasat, Kolkata, 700125, West Bengal, India
| | - Preity Pragnya Sahoo
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Laxmi Narayan Tiwari
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Basant Narain Singh
- Department of Botany, Pandit Deendayal Upadhyaya Shekhawati University, Sikar, Nawalgarh Road, Katrathal, Rajasthan, 332024, India
| | - Rashmi Kumari
- Department of Zoology, ZA Islamia College Siwan, Affiliated Unit of Jai Prakash University, Chapra, Bihar, 841226, India
| | - Anisha Kumari
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India
| | - Ankit Rai
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India.
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India.
| |
Collapse
|
204
|
Oli AN, Adejumo SA, Rowaiye AB, Ogidigo JO, Hampton-Marcell J, Ibeanu GC. Tumour Immunotherapy and Applications of Immunological Products: A Review of Literature. J Immunol Res 2024; 2024:8481761. [PMID: 39483536 PMCID: PMC11527548 DOI: 10.1155/2024/8481761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 11/03/2024] Open
Abstract
Malignant tumors, characterized by uncontrolled cell proliferation, are a leading global health challenge, responsible for over 9.7 million deaths in 2022, with new cases expected to rise to 35 million annually by 2050. Immunotherapy is preferred to other cancer therapies, offering precise targeting of malignant cells while simultaneously strengthening the immune system's complex responses. Advances in this novel field of science have been closely linked to a deeper knowledge of tumor biology, particularly the intricate interplay between tumor cells, the immune system, and the tumor microenvironment (TME), which are central to cancer progression and immune evasion. This review offers a comprehensive analysis of the molecular mechanisms that govern these interactions, emphasizing their critical role in the development of effective immunotherapeutic products. We critically evaluate the current immunotherapy approaches, including cancer vaccines, adoptive T cell therapies, and cytokine-based treatments, highlighting their efficacy and safety. We also explore the latest advancements in combination therapies, which synergistically integrate multiple immunotherapeutic strategies to overcome resistance and enhance therapeutic outcomes. This review offers key insights into the future of cancer immunotherapy with a focus on advancing more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Angus Nnamdi Oli
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420211, Nigeria
| | - Samson Adedeji Adejumo
- Department of Biological Sciences, University of Illinois, Chicago, 845 West Taylor, Chicago 60607, Illinois, USA
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Federal University Oye Ekiti, Oye, Ekiti State, Nigeria
| | - Adekunle Babajide Rowaiye
- National Biotechnology Development Agency, Abuja 900211, Nigeria
- Department of Pharmaceutical Science, North Carolina Central University, Durham 27707, North Carolina, USA
| | | | - Jarrad Hampton-Marcell
- Department of Biological Sciences, University of Illinois, Chicago, 845 West Taylor, Chicago 60607, Illinois, USA
| | - Gordon C. Ibeanu
- Department of Pharmaceutical Science, North Carolina Central University, Durham 27707, North Carolina, USA
| |
Collapse
|
205
|
Kumar Y, Shrivastav S, Garg K, Modi N, Wiltos K, Woźniak M, Ijaz MF. Automating cancer diagnosis using advanced deep learning techniques for multi-cancer image classification. Sci Rep 2024; 14:25006. [PMID: 39443621 PMCID: PMC11499884 DOI: 10.1038/s41598-024-75876-2] [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: 08/27/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Cancer detection poses a significant challenge for researchers and clinical experts due to its status as the leading cause of global mortality. Early detection is crucial, but traditional cancer detection methods often rely on invasive procedures and time-consuming analyses, creating a demand for more efficient and accurate solutions. This paper addresses these challenges by utilizing automated cancer detection through AI-based techniques, specifically focusing on deep learning models. Convolutional Neural Networks (CNNs), including DenseNet121, DenseNet201, Xception, InceptionV3, MobileNetV2, NASNetLarge, NASNetMobile, InceptionResNetV2, VGG19, and ResNet152V2, are evaluated on image datasets for seven types of cancer: brain, oral, breast, kidney, Acute Lymphocytic Leukemia, lung and colon, and cervical cancer. Initially, images undergo segmentation techniques, proceeded by contour feature extraction where parameters such as perimeter, area, and epsilon are computed. The models are rigorously evaluated, with DenseNet121 achieving the highest validation accuracy as 99.94%, 0.0017 as loss, and the lowest Root Mean Square Error (RMSE) values as 0.036056 for training and 0.045826 for validation. These results revealed the capability of AI-based techniques in improving cancer detection accuracy, with DenseNet121 emerging as the most effective model in this study.
Collapse
Affiliation(s)
- Yogesh Kumar
- Department of Computer Science and Engineering, School of Technology, PDEU, Gandhinagar, Gujarat, 382426, India
| | | | - Kinny Garg
- Department of ECE, AMC Engineering College, Bengaluru, Karnataka, India
| | - Nandini Modi
- Department of Computer Science and Engineering, School of Technology, PDEU, Gandhinagar, Gujarat, 382426, India.
| | - Katarzyna Wiltos
- Faculty of Applied Mathematics, Silesian University of Technology, Kaszubska 23, Gliwice, 44100, Poland
| | - Marcin Woźniak
- Faculty of Applied Mathematics, Silesian University of Technology, Kaszubska 23, Gliwice, 44100, Poland.
| | - Muhammad Fazal Ijaz
- School of IT and Engineering, Melbourne Institute of Technology, Melbourne, 3000, Australia.
| |
Collapse
|
206
|
Ojedele OA, Umar HI, Baammi S, Metouekel A, Mengistie AA, Bin Jardan YA, Shazly GA, Victor O. Cheminformatics-aided discovery of potential allosteric site modulators of ubiquitin-specific protease 7. Sci Rep 2024; 14:24995. [PMID: 39443474 PMCID: PMC11499889 DOI: 10.1038/s41598-024-74851-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Ubiquitin-specific peptidase 7 (USP7) is a deubiquitinating enzyme that mediates the stability and activity of numerous proteins. At basal expression levels, USP7 stabilizes p53 protein, even in the presence of excess MDM2. However, its overexpression leads to the deubiquitination of MDM2 at a rate faster than p53, leading to p53 degradation and pro-tumorigenic roles. Consequently, it is an attractive target for anticancer drug discovery via the modulation of its allosteric site from which the protein is activated. In this study, molecular modeling techniques and cheminformatics approaches were employed to unravel the potential of eighty compounds to serve as its allosteric site modulators. The compounds were initially subjected to virtual screening. Subsequently, the binding free energies of the top four compounds with the highest binding affinities were calculated, and their drug-likeness, and pharmacokinetic and toxicity profiles were evaluated. Ultimately, the complexes of the protein and hit compounds were subjected to a 100 nanoseconds (ns) molecular dynamics simulation. The results of the study revealed eight compounds from the compound library with docking scores ranging from - 7.491 to -11.43 kcal/mol, compared to P217564, which exhibited a docking score of -5.671 kcal/mol. The top four compounds with the highest affinities possessed drug-like properties, and good pharmacokinetic and toxicity profiles, and their predicted inhibitory potentials showed they will be effective at minimal concentration. Also, molecular dynamics simulation confirmed the stability of the protein-ligand complexes. Conclusively, the compounds identified in this study are worthy of further evaluation for the development of allosteric site modulators of USP7.
Collapse
Affiliation(s)
- Olayinka Abraham Ojedele
- Department of Biochemistry, School of Life Sciences (SLS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria
- Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria
| | - Haruna Isiyaku Umar
- Department of Biochemistry, School of Life Sciences (SLS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria
- Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria
| | - Soukayna Baammi
- Bioinformatics Laboratory, College of Computing, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Amira Metouekel
- University of Technology of Compiègne, EA 4297 TIMR, Compiègne Cedex, 60205, France
| | | | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 11451, Riyadh, 11451, Saudi Arabia
| | - Gamal A Shazly
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 11451, Riyadh, 11451, Saudi Arabia
| | - Omoboyede Victor
- Department of Biochemistry, School of Life Sciences (SLS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
- Computer Aided Therapeutics and Drug Design (CATDD) Group, School of Sciences (SOS), Federal University of Technology Akure, P.M.B 704, Akure, Nigeria.
| |
Collapse
|
207
|
Karami F, Osanloo M, Alipanah H, Zarenezhad E, Moghimi F, Ghanbariasad A. Comparison of the efficacy of alginate nanoparticles containing Cymbopogon citratus essential oil and citral on melanoma and breast cancer cell lines under normoxic and hypoxic conditions. BMC Complement Med Ther 2024; 24:372. [PMID: 39427126 PMCID: PMC11490153 DOI: 10.1186/s12906-024-04673-1] [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: 07/02/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Solid tumors often develop hypoxic regions, leading to aggressive behavior and increased drug resistance. METHODS The chemical composition of Cymbopogon citratus essential oil (EO) was analyzed using GC-MS. Alginate nanoparticles containing the EO and its primary component, citral, were synthesized via the ionic gelation method. Encapsulation was confirmed using ATR-FTIR analysis. The anticancer efficacy of C. citratus EO, citral, and their respective alginate nanoparticles was evaluated under normoxic (21% oxygen) and hypoxic (1% oxygen) conditions on breast cancer (MDA-MB-231) and melanoma (A-375) cell lines. Additionally, qPCR and flow cytometry were used to assess apoptosis gene expression ratios (Bax/Bcl-2) and levels of apoptosis. RESULTS Citral (80.98%) was identified as the major component of the EO. Alginate nanoparticles containing C. citratus EO and citral (C. citratus-AlgNPs and citral-AlgNPs) were synthesized with particle sizes of 195 ± 4 nm and 222 ± 9 nm, and zeta potentials of -22 ± 3 mV and - 17 ± 1 mV, respectively. Both samples demonstrated significantly greater efficacy under hypoxic conditions. Citral and C. citratus-AlgNPs had IC50 values of 27 (19-39) µg/mL and 25 (4-147) µg/mL, respectively, against MDA-MB-231 and A-375 cells. Flow cytometry showed increased apoptosis under hypoxic conditions, with the highest rates observed for citral-AlgNPs and C. citratus-AlgNPs (84 ± 5 and 92 ± 5% in MDA-MB-231 and A-375 cells, respectively). CONCLUSION This study demonstrates that alginate nanoparticles enhance the anticancer activity of C. citratus-AlgNPs and citral, particularly under hypoxic conditions, highlighting their potential for hypoxia-targeted cancer therapies.
Collapse
Affiliation(s)
- Farnaz Karami
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahmoud Osanloo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Hiva Alipanah
- Department of Physiology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Fatemeh Moghimi
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
208
|
Moralev AD, Salomatina OV, Salakhutdinov NF, Zenkova MA, Markov AV. Soloxolone N-3-(Dimethylamino)propylamide Restores Drug Sensitivity of Tumor Cells with Multidrug-Resistant Phenotype via Inhibition of P-Glycoprotein Efflux Function. Molecules 2024; 29:4939. [PMID: 39459307 PMCID: PMC11510211 DOI: 10.3390/molecules29204939] [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: 09/17/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Multidrug resistance (MDR) remains a significant challenge in cancer therapy, primarily due to the overexpression of transmembrane drug transporters, with P-glycoprotein (P-gp) being a central focus. Consequently, the development of P-gp inhibitors has emerged as a promising strategy to combat MDR. Given the P-gp targeting potential of soloxolone amides previously predicted by us by an absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis, the aim of the current study was to experimentally verify their P-gp inhibitory and MDR reversing activities in vitro. Screening of soloxolone amides as modulators of P-gp using molecular docking and cellular P-gp substrate efflux assays revealed the ability of compound 4 bearing a N-3-(dimethylamino)propylamide group to interact with the active site of P-gp and inhibit its transport function. Blind and site-specific molecular docking accompanied by a kinetic assay showed that 4 directly binds to the P-gp transmembrane domain with a binding energy similar to that of zosuquidar, a third-generation P-gp inhibitor (ΔG = -10.3 kcal/mol). In vitro assays confirmed that compound 4 enhanced the uptake of Rhodamine 123 (Rho123) and doxorubicin (DOX) by the P-gp-overexpressing human cervical carcinoma KB-8-5 (by 10.2- and 1.5-fold, respectively (p < 0.05, unpaired t-test)) and murine lymphosarcoma RLS40 (by 15.6- and 1.75-fold, respectively (p < 0.05, unpaired t-test)) cells at non-toxic concentrations. In these cell models, 4 showed comparable or slightly higher activity than the reference inhibitor verapamil (VPM), with the most pronounced effect of the hit compound in Rho123-loaded RLS40 cells, where 4 was 2-fold more effective than VPM. Moreover, 4 synergistically restored the sensitivity of KB-8-5 cells to the cytotoxic effect of DOX, demonstrating MDR reversal activity. Based on the data obtained, 4 can be considered as a drug candidate to combat the P-gp-mediated MDR of tumor cells and semisynthetic triterpenoids, with amide moieties in general representing a promising scaffold for the development of novel therapeutics for tumors with low susceptibility to antineoplastic agents.
Collapse
Affiliation(s)
- Arseny D. Moralev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.D.M.); (O.V.S.); (M.A.Z.)
| | - Oksana V. Salomatina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.D.M.); (O.V.S.); (M.A.Z.)
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Nariman F. Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.D.M.); (O.V.S.); (M.A.Z.)
| | - Andrey V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.D.M.); (O.V.S.); (M.A.Z.)
| |
Collapse
|
209
|
Al-Wahaibi LH, El-Sheref EM, Tawfeek HN, Abou-Zied HA, Rabea SM, Bräse S, Youssif BGM. Design, synthesis, and biological evaluation of novel quinoline-based EGFR/HER-2 dual-target inhibitors as potential anti-tumor agents. RSC Adv 2024; 14:32978-32991. [PMID: 39434991 PMCID: PMC11492426 DOI: 10.1039/d4ra06394e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Dual targeting of EGFR and HER2 is a valid anti-cancer approach for treating solid tumors. We designed and synthesized a new series of EGFR/HER-2 dual-target inhibitors based on quinoline derivatives. The structure of the newly synthesized compounds was verified using 1H NMR, 13C NMR, and elemental analysis. The targeted compounds were tested for antiproliferative efficacy against four cancer cell lines. All the compounds had GI50s ranging from 25 to 82 nM, with breast (MCF-7) and lung (A-549) cancer cell lines being the most sensitive. Compound 5a demonstrated the most significant antiproliferative action. With inhibitory (IC50) values of 71 and 31 nM, respectively, compound 5a proved to be the most effective dual-target inhibitor of EGFR and HER-2, outperforming the reference erlotinib (IC50 = 80 nM) as an EGFR inhibitor but falling short of the clinically used agent lapatinib (IC50 = 26 nM) as a HER2 inhibitor. The apoptotic potential activity of 5a was examined, and the findings demonstrated that 5a promotes apoptosis by activating caspase-3, 8, and Bax while simultaneously reducing the expression of the anti-apoptotic protein Bcl-2. The docking studies provided valuable insights into the binding interactions of compounds 3e and 5a with EGFR, effectively rationalizing the observed SAR trends.
Collapse
Affiliation(s)
- Lamya H Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah Bint Abdulrahman University Riyadh 11671 Saudi Arabia
| | - Essmat M El-Sheref
- Chemistry Department, Faculty of Science, Minia University El Minia 61519 Egypt
| | - Hendawy N Tawfeek
- Chemistry Department, Faculty of Science, Minia University El Minia 61519 Egypt
- Unit of Occupational of Safety and Health, Administration Office of Minia University El-Minia 61519 Egypt
| | - Hesham A Abou-Zied
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University Minia Egypt
| | - Safwat M Rabea
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University Minia 61519 Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology Karlsruhe 76131 Germany
| | - Bahaa G M Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt +20-01098294419
| |
Collapse
|
210
|
Ensoy M, Cansaran-Duman D. Targeting Ferroptosis with Small Molecule Atranorin (ATR) as a Novel Therapeutic Strategy and Providing New Insight into the Treatment of Breast Cancer. Pharmaceuticals (Basel) 2024; 17:1380. [PMID: 39459017 PMCID: PMC11509994 DOI: 10.3390/ph17101380] [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: 08/06/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Ferroptosis results from the accumulation of iron-dependent lipid peroxides and reactive oxygen species (ROS). Previous research has determined the effect of atranorin (ATR) on other cell death mechanisms, but its potential for a ferroptotic effect depending on ROS levels is unclear. This study details the therapeutic role of small-molecule ATR through ferroptosis by suppressing MDA-MB-231, MCF-7, BT-474, and SK-BR-3 breast cancer cells. Methods: The anti-proliferative effect of ATR on cells was evaluated by xCELLigence analysis, and ferroptotic activity was evaluated by enzymatic assay kits. The changes in gene and protein expression levels of ATR were investigated by the qRT-PCR and western blot. In addition, mitochondrial changes were examined by transmission electron microscopy. Results: ATR was found to reduce cell viability in cancer cells in a dose- and time-dependent manner without showing cytotoxic effects on normal breast cells. In BT-474 and MDA-MB-231 cells, ATR, which had a higher anti-proliferative effect, increased iron, lipid peroxidation, and ROS levels in cells and decreased the T-GSH/GSSG ratio. The results revealed for the first time that small-molecule ATR exhibited anti-cancer activity by inducing the glutathione pathway and ferroptosis. Conclusions: This study highlights the potential of ATR as a drug candidate molecule that can be used in the development of new therapeutic strategies for the treatment of triple-negative and luminal-B breast cancer subtypes.
Collapse
|
211
|
Kalaimani K, Balachandran S, Boopathy LK, Roy A, Jayachandran B, Sankaranarayanan S, Arumugam MK. Recent advancements in small interfering RNA based therapeutic approach on breast cancer. Eur J Pharmacol 2024; 981:176877. [PMID: 39128807 DOI: 10.1016/j.ejphar.2024.176877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Breast cancer (BC) is the most common and malignant tumor diagnosed in women, with 2.9 million cases in 2023 and the fifth highest cancer-causing mortality worldwide. Recent developments in targeted therapy options for BC have demonstrated the promising potential of small interfering RNA (siRNA)-based cancer therapeutic approaches. As BC continues to be a global burden, siRNA therapy emerges as a potential treatment strategy to regulate disease-related genes in other types of cancers, including BC. siRNAs are tiny RNA molecules that, by preventing their expression, can specifically silence genes linked to the development of cancer. In order to increase the stability and effectiveness of siRNA delivery to BC cells, minimize off-target effects, and improve treatment efficacy, advanced delivery technologies such as lipid nanoparticles and nanocarriers have been created. Additionally, combination therapies, such as siRNAs that target multiple pathways are used in conjunction with conventional chemotherapy agents, have shown synergistic effects in various preclinical studies, opening up new treatment options for breast cancer that are personalized and precision medicine-oriented. Targeting important genes linked to BC growth, metastasis, and chemo-resistance has been reported in BC research using siRNA-based therapies. This study reviews recent reports on therapeutic approaches to siRNA for advanced treatment of BC. Furthermore, this review evaluates the role and mechanisms of siRNA in BC and demonstrates the potential of exploiting siRNA as a novel target for BC therapy.
Collapse
Affiliation(s)
- Kathirvel Kalaimani
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Shana Balachandran
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Lokesh Kumar Boopathy
- Centre for Laboratory Animal Technology and Research, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Anitha Roy
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India
| | - Bhuvaneshwari Jayachandran
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Sangamithra Sankaranarayanan
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Madan Kumar Arumugam
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India.
| |
Collapse
|
212
|
Bérot A, Maniti O, El Alaoui S, Granjon T, El Alaoui M. Generation of Anti-Epidermal Growth Factor Receptor-2 (HER2) Immunoliposomes Using Microbial Transglutaminase (mTG)-Mediated Site-Specific Conjugated Antibodies. ACS Pharmacol Transl Sci 2024; 7:3034-3044. [PMID: 39416960 PMCID: PMC11475288 DOI: 10.1021/acsptsci.4c00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
Nanocarriers have found their interests in many fields including drug delivery and labeling of cells with the aim to target and eradicate tumor cells. One of the approaches to specifically address nanocarriers, such as liposomes, to their target is to attach antibodies of interest to their surface. To date, the development of immunoliposomes has been widely explored but has mainly involved chemical and unspecific reactions that could impair antibody stability, integrity, and orientation, thus reducing optimized immunoliposomes generation. In this study, we report the use of the patented COVISOLINK technology and the strain-promoted alkyne-azide cycloaddition (SPAAC) to generate immunoliposomes that target HER2 positive breast cancer with Trastuzumab as the antibody to be coupled. The efficacy of our two-step functionalization strategy and the successful specific coupling of the antibodies were validated by high-performance liquid chromatography-size exclusion chromatography (HPLC-SEC), which allowed a precise quantification of antibodies conjugated to liposomes and confirmed by cryo-TEM and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analyses. We also demonstrate by flow cytometry and epifluorescence microscopy that the produced anti-HER2 immunoliposomes were able to interact specifically with their target cells (SK-BR-3) while remaining negative with cells that express HER2 at a low level (MDA-MB-231). Hence, for the first time, our COVISOLINK strategy using microbial transglutaminase (mTG) enables the preparation and production of well-characterized immunoliposomes that could be used in different applications, including therapies.
Collapse
Affiliation(s)
- Anna Bérot
- Covalab, 1B Rue Jacques Monod, 69500 Bron, France
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires
ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | - Ofelia Maniti
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires
ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | | | - Thierry Granjon
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires
ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | | |
Collapse
|
213
|
Attar GS, Kumar M, Bhalla V. Targeting sub-cellular organelles for boosting precision photodynamic therapy. Chem Commun (Camb) 2024; 60:11610-11624. [PMID: 39320942 DOI: 10.1039/d4cc02702g] [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: 09/26/2024]
Abstract
Among various cancer treatment methods, photodynamic therapy has received significant attention due to its non-invasiveness and high efficiency in inhibiting tumour growth. Recently, specific organelle targeting photosensitizers have received increasing interest due to their precise accumulation and ability to trigger organelle-mediated cell death signalling pathways, which greatly reduces the drug dosage, minimizes toxicity, avoids multidrug resistance, and prevents recurrence. In this review, recent advances and representative photosensitizers used in targeted photodynamic therapy on organelles, specifically including the endoplasmic reticulum, Golgi apparatus, mitochondria, nucleus, and lysosomes, have been comprehensively reviewed with a focus on organelle structure and organelle-mediated cell death signalling pathways. Furthermore, a perspective on future research and potential challenges in precision photodynamic therapy has been presented at the end.
Collapse
Affiliation(s)
- Gopal Singh Attar
- Department of chemistry UGC Sponsored-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar-143005, Punjab, India.
| | - Manoj Kumar
- Department of chemistry UGC Sponsored-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar-143005, Punjab, India.
| | - Vandana Bhalla
- Department of chemistry UGC Sponsored-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar-143005, Punjab, India.
| |
Collapse
|
214
|
Parkin HC, Shopperly LK, Perez MR, Willerth SM, Manners I. Uniform block copolymer nanofibers for the delivery of paclitaxel in 2D and 3D glioblastoma tumor models. Biomater Sci 2024; 12:5283-5294. [PMID: 39246052 DOI: 10.1039/d4bm00480a] [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: 09/10/2024]
Abstract
Cancer treatment has transformed in recent years, with the introduction of immunotherapy providing substantial improvements in prognoses for certain cancers. However, traditional small molecule chemotherapeutics remain the major frontline of defence, and improving their delivery to solid tumors is of utmost importance for improving potency and reducing side effects. Here, length-controlled one-dimensional seed nanofibers (ca. 25 nm, ĐL = 1.05) were generated from poly(fluorenetrimethylenecarbonate)-block-poly(dimethylaminoethylmethacrylate) via living crystallization-driven self-assembly. Paclitaxel, with an encapsulation content ranging from 1 to 100 wt%, was loaded onto the preformed nanoparticles by solvent addition and evaporation. Drug loading was quantified by dynamic light scattering and transmission electron microscopy. Drug-loaded vectors were then incubated with U87 MG glioblastoma cells in a 2D cell assay for up to 72 h, and their anticancer properties were determined. It was observed that seed nanofibers loaded with 20 wt% paclitaxel were the most advantageous combination (IC50 = 0.48 μg mL-1), while pure seed nanofibers with no loaded drug displayed much lower cytotoxicity (IC50 = 11.52 μg mL-1). The IC50 of the loaded seed nanofibers rivaled that of the commercially approved Abraxane® (IC50 = 0.46 μg mL-1). 3D tumor spheroids were then cultured and subjected to the same stresses. Live/dead cell staining revealed that once more, seed nanofibers with 20 wt% paclitaxel, Abraxane®, and paclitaxel all exhibited similar levels of potency (55% viability), whereas control samples exhibited much higher cell viability (70%) after 3 days. These results demonstrate that nanofibers contain great potential as biocompatible drug delivery vehicles for cancer treatment as they exert a similar anticancer effect to the commercially available Abraxane®.
Collapse
Affiliation(s)
- Hayley C Parkin
- Department of Chemistry, University of Victoria, Victoria, BC V8 W 3 V6, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.
| | - Lennard K Shopperly
- Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, BC V8 W 3 V6, Canada
| | - Milena R Perez
- Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, BC V8 W 3 V6, Canada
| | - Stephanie M Willerth
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.
- Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, BC V8 W 3 V6, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Ian Manners
- Department of Chemistry, University of Victoria, Victoria, BC V8 W 3 V6, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.
| |
Collapse
|
215
|
Scorzo AV, Byrd BK, Kwon CY, Strawbridge RR, Samkoe KS, Hoopes PJ, Paulsen KD, Roberts DW, Davis SC. Whole-body fluorescence cryotomography identifies a fast-acting, high-contrast, durable contrast agent for fluorescence-guided surgery. Theranostics 2024; 14:6426-6445. [PMID: 39479457 PMCID: PMC11519800 DOI: 10.7150/thno.100802] [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: 07/11/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024] Open
Abstract
Imaging of tumor-specific fluorescent contrast agents to guide tumor removal has been shown to improve outcomes and is now standard practice for some neurosurgical procedures. However, many agents require administration hours before surgery, a practical challenge, and may exhibit inconsistent concordance with contrast-enhanced MRI (CE-MRI), the current standard for diagnosing and guiding glioma removal. A fluorescent agent that accurately marks tumor shortly after administration and is otherwise similar to CE-MRI would help overcome these shortcomings. Methods: We used whole-body 3-D fluorescence cryo-imaging and co-registered CE-MRI volumes to evaluate several fluorescent contrast agent candidates for diagnostic performance and concordance with CE-MRI. Mice with brain tumors were administered a cocktail of fluorescent agent candidates and a MRI contrast agent, and then imaged with MRI and fluorescence cryo-imaging at several timepoints after administration. The high-resolution 3-D cryo-imaging volumes of the fluorescent agents were used to determine diagnostic performance metrics and correlation with CE-MRI. Results: While all agents showed positive metrics, one agent, tetramethylrhodamine conjugated to a small polyethylene glycol chain (TMR-PEG1k), outperformed the others, exhibiting minimal normal brain signal, high tumor-to-background-ratio, diagnostic accuracy, and cross-correlation to CE-MRI at all post-administration timepoints (10-90 min) and tumor lines examined. Conclusion: These favorable properties establish TMR-PEG1k as a promising candidate for surgical guidance.
Collapse
Affiliation(s)
- Augustino V. Scorzo
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Brook K. Byrd
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Caleb Y. Kwon
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Kimberley S. Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - P. Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - David W. Roberts
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Scott C. Davis
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
216
|
Lan T, He S, Luo X, Pi Z, Lai W, Jiang C, Gan J, Wei S, Wu Z, Yun C, Leng J, Li C. Disruption of NADPH homeostasis by total flavonoids from Adinandra nitida Merr. ex Li leaves triggers ROS-dependent p53 activation leading to apoptosis in non-small cell lung cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118340. [PMID: 38762212 DOI: 10.1016/j.jep.2024.118340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/03/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Adinandra nitida Merr. ex Li leaves serve as a herbal tea and hold a significant role in traditional Chinese medicine, being applied to assist in tumor treatment. Flavonoids present the primary bioactive constituents in Adinandra nitida Merr. ex Li leaves. AIM OF THE STUDY To explore the potential of total flavonoids from Adinandra nitida Merr. ex Li Leaves (TFAN) in inhibiting non-small cell lung cancer (NSCLC) and further elucidate the underlying mechanisms. MATERIALS AND METHODS Human NSCLC cell lines and normal lung cell line were employed to assess the impact of TFAN (0-160 μg/mL for 24, 28 and 72 h) on cell proliferation in vitro. Immunofluorescence (IF) staining gauged p53 expression changes in NSCLC cells under TFAN present condition (150 μg/mL for 24 h). In vivo study utilized NSCLC cell derived xenograft tumors in nude mice, administering TFAN orally (200 and 400 mg/kg) for 14 days. Immunohistochemistry assessed Cleaved Caspase 3 expression change in A549 xenograft tumors treated with TFAN (400 mg/kg for 14 days). RNA-seq and KEGG analysis identified gene expression changes and enriched processes in A549 xenograft tumors treated with TFAN. CM-H2DCFDA and metabolomics assessed ROS level and GSH/GSSG pool changes in A549 cells under TFAN present condition. Cell viability assay and IF staining assessed A549 cell proliferation and p53 expression changes under H2O2-induced oxidative stress (0-40 μM for 24 h) and TFAN present conditions. GSEA and N-Acetyl-L-cysteine (NAC) rescue (0-1 μM for 24 h) analyzed the impact of TFAN on GSH de novo synthesis. NADPH/NADP+ pool measurement and NADPH rescue (0-10 μM for 24 h) analyzed the impact of TFAN on GSH salvage synthesis. GC-FID and HPLC-MS were utilized to detect ethanol and ethyl acetate residues, and to characterize the chemical constituents in TFAN, respectively. The total flavonoid content of TFAN was determined using a 330 nm wavelength. RESULTS TFAN significantly inhibited A549 cells (wild-type p53) but not NCI-H1299 cells (p53-deficient), NCI-H596 cells (p53-mutant) or BEAS-2B in vitro. IF staining validated p53 genotype for the cell lines and revealed an increase in p53 expression in A549 cells after TFAN treatment. In vivo, TFAN selectively inhibited A549 xenograft tumor growth without discernible toxicity, inducing apoptosis evidenced by Cleaved Caspase 3 upregulation. RNA-seq and KEGG analysis suggested ROS biosynthesis was involved in TFAN-induced p53 activation in A549 cells. Elevated ROS level in TFAN-treated A549 cells were observed. Moreover, TFAN sensitized A549 cells to H2O2-induced oxidative stress, with higher p53 expression. Additionally, A549 cells compensated with GSH de novo synthesis under TFAN present condition, confirmed by GSEA and NAC rescue experiment. TFAN disrupted NADPH homeostasis to impair GSH salvage biosynthesis, supported by NADPH/NADP+ change and NADPH rescue experiment. The chemical constituents of TFAN, with acceptable limits for ethanol and ethyl acetate residues and a total flavonoid content of 68.87%, included Catechin, Epicatechin, Quercitroside, Camellianin A, and Apigenin. CONCLUSION The disruption of NADPH homeostasis by TFAN triggers ROS-dependent p53 activation that leads to apoptotic cell death, ultimately suppressing NSCLC growth. These findings offer potential therapeutic implications of Adinandra nitida Merr. ex Li leaves in combating NSCLC.
Collapse
Affiliation(s)
- Taijin Lan
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; Key Laboratory of Integrative Translational Medicine of Guangxi High Incidence Infectious Diseases, Nanning 530200, China; School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Songhua He
- Guangxi Institute for Food and Drug Control, Nanning 530021, China
| | - Xuefei Luo
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhenyu Pi
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Weihui Lai
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Chunhui Jiang
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jun Gan
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Suyun Wei
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhanshuai Wu
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Chenxia Yun
- Key Laboratory of Integrative Translational Medicine of Guangxi High Incidence Infectious Diseases, Nanning 530200, China.
| | - Jing Leng
- Key Laboratory of Integrative Translational Medicine of Guangxi High Incidence Infectious Diseases, Nanning 530200, China.
| | - Changlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
217
|
Morgan NR, Ramdas P, Bhuvanendran S, Radhakrishnan AK. Delineating the Immunotherapeutic Potential of Vitamin E and Its Analogues in Cancer: A Comprehensive Narrative Review. BIOMED RESEARCH INTERNATIONAL 2024; 2024:5512422. [PMID: 39416707 PMCID: PMC11480965 DOI: 10.1155/2024/5512422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024]
Abstract
Cancer is a disease resulting from uncontrolled cell division, which significantly contributes to human mortality rates. An alternative approach to cancer treatment, such as cancer immunotherapy, is needed as the existing chemotherapy and radiotherapy approaches target the cancer cells and healthy dividing cells. Vitamin E is a plant-derived lipid-soluble antioxidant with numerous health-promoting benefits, including anticancer and immunomodulatory properties. Vitamin E comprises eight natural isoforms: tocopherols (α, β, δ, and γ) and tocotrienols (α, β, δ, and γ). While initial research focused on the anticancer properties of α-tocopherol, there is growing interest in other natural forms and modified synthetic analogues of vitamin E due to their unique properties and enhanced anticancer effects. Hence, this review is aimed at outlining the effect of vitamin E and its analogues at various steps of the cancer-immunity cycle that can be used to stimulate anticancer immune responses.
Collapse
Affiliation(s)
- Nevvin Raaj Morgan
- Food as Medicine Research StrengthJeffrey Cheah School of Medicine and Health SciencesMonash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Premdass Ramdas
- Food as Medicine Research StrengthJeffrey Cheah School of Medicine and Health SciencesMonash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Saatheeyavaane Bhuvanendran
- Food as Medicine Research StrengthJeffrey Cheah School of Medicine and Health SciencesMonash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Ammu Kutty Radhakrishnan
- Food as Medicine Research StrengthJeffrey Cheah School of Medicine and Health SciencesMonash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| |
Collapse
|
218
|
Peng Y, Hu Y, Liu Y, Lin H. Green Synthesis of Dracocephalum kotschyi-Coated Silver Nanoparticles: Antimicrobial, Antioxidant, and Anticancer Potentials. Med Sci Monit 2024; 30:e944823. [PMID: 39358918 PMCID: PMC11460312 DOI: 10.12659/msm.944823] [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: 04/15/2024] [Accepted: 08/08/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The environmentally friendly production of silver nanoparticles (AgNPs) has gained significant attention as a sustainable alternative to traditional chemical methods. This study focused on synthesizing AgNPs using extract of Dracocephalum kotschyi (D. kotschyi), a medicinal plant. MATERIAL AND METHODS The biosynthesis of AgNPs was monitored using UV-visible spectrophotometry. The role of phytoconstituents from D. kotschyi in stabilizing AgNPs was analyzed using Fourier-transform infrared (FTIR) spectroscopy. Dynamic light scattering (DLS) spectroscopy was used to determine the size, charge, and polydispersity of the nanoparticles, while scanning electron microscopy (SEM) was employed to assess their morphology. We evaluated the antimicrobial efficacy of the synthesized AgNPs against various bacteria, their antioxidant properties via a 2,2-Diphenyl-1-picrylhydrazyl (DPPH) assay, and their cytotoxic activity against the HeLa cervical cancer cell line. RESULTS The formation of AgNPs was indicated by a color change and the emergence of a surface plasmon resonance peak at 418 nm. The nanoparticles demonstrated significant antimicrobial, antioxidant, cytotoxic, and anticancer activities. Morphology, size, and shape analysis revealed nearly spherical particles with an average size of 43 nm. FTIR confirmed the presence of phenolic compounds in the extract, serving as reducing and capping agents. X-ray diffraction (XRD) analysis confirmed the crystalline structure of the nanoparticles. Antimicrobial assessments showed effectiveness against Escherichia coli and Staphylococcus aureus. The DPPH scavenging assay demonstrated efficient antioxidant activity, and potent apoptotic anticancer effects were observed on cervical cancer cells. CONCLUSIONS The extract of D. kotschyi was effective as a reducing agent in the environmentally friendly synthesis of AgNPs, which exhibited noteworthy antimicrobial, antioxidant, and anticancer properties. These findings suggest potential biomedical applications for the synthesized AgNPs.
Collapse
Affiliation(s)
- Yibo Peng
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, Zhejiang, PR China
| | - Ying Hu
- Department of Pharmacy, Zhejiang Pharmaceutical University, Nibo, Zhejiang, PR China
| | - Yang Liu
- Department of Pharmacy, Hunan Province Directly Affiliated TCM Hospital, Zhuzhou, Hunan, PR China
| | - Hangjuan Lin
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, Zhejiang, PR China
| |
Collapse
|
219
|
Thozhukkad Moosaripparambil S, Vadakkadath Meethal K. Unveiling the anticancer potential of Anamirta cocculus (L.) Wight& Arn.: Evidences from cytotoxicity studies, apoptosis analysis, and molecular docking. 3 Biotech 2024; 14:245. [PMID: 39345961 PMCID: PMC11424601 DOI: 10.1007/s13205-024-04096-2] [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: 05/25/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Anamirta cocculus, a woody climber, is extensively utilised in traditional Asian medicine. This study investigates the cytotoxic effects of A. cocculus leaf extracts on various cancer cell lines as well as on a normal cell line. The ethyl acetate extract exhibited potent anticancer activity, with the highest cytotoxicity observed against ovarian cancer cell line (PA1) (IC50 = 8.30 ± 0.38 µg/mL) and colorectal adenocarcinoma cell line (HT29) (IC50 = 17.97 ± 0.63 µg/mL). Notably, the extract displayed low toxicity (18.72 ± 0.73%) on the normal human keratinocyte cell line (HaCaT) at a concentration of 100 µg/mL, indicating selective cytotoxicity towards cancer cells. The acetone extract also demonstrated significant cytotoxicity against various cancer cell lines, including A498, MG63, PA1, and UM-SCC-83B. The ethyl acetate extract of A. cocculus demonstrated potent inhibition of colony formation in HT29 and PA1 cancer cell lines while inducing apoptosis, as evidenced by membrane blebbing, chromatin condensation, and DNA fragmentation. The number of late apoptotic cells increased with an increase in concentrations of ACLE. Molecular docking studies of compounds identified through GC-MS analysis revealed strong interactions with key apoptotic proteins, including caspase-8, p53, caspase-3, and caspase-9. Compounds such as vitamin E, epoxylathyrol, squalene, and phytol showed high binding affinity to these proteins, suggesting their role in apoptosis induction. The possibility of induction of apoptotic proteins through indirect interaction by binding to other proteins or receptors cannot be ruled out. The cytotoxic effects may result from individual, combined, or synergistic actions of these compounds. Among these, epoxylathyrol emerged as a particularly promising anticancer drug candidate based on ADME analysis and binding affinity assessments, warranting further investigation. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04096-2.
Collapse
Affiliation(s)
| | - Kannan Vadakkadath Meethal
- Department of Zoology, University of Calicut, Calicut University PO, Malappuram District, Malappuram, Kerala 673635 India
- Centre for Advances in Molecular Biology, University of Calicut, Calicut University PO, Malappuram District, Malappuram, Kerala 673635 India
| |
Collapse
|
220
|
Almujri SS, Almalki WH. The paradox of autophagy in cancer: NEAT1's role in tumorigenesis and therapeutic resistance. Pathol Res Pract 2024; 262:155523. [PMID: 39173466 DOI: 10.1016/j.prp.2024.155523] [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: 04/24/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
Cancer remains a current active problem of modern medicine, a process during which cell growth and proliferation become uncontrolled. However, the role of autophagy in the oncological processes is counterintuitive and, at the same time, increasingly influential on the formation, development, and response to therapy of oncological diseases. Autophagy is a vital cellular process that removes defective proteins and organelles and supports cellular homeostasis. Autophagy can enhance the ability to form new tumors and suppress this formation in cancer. The dual potential of apoptosis may be the reason for this duality in either promoting or impeding the survival of cancer cells, depending on the situation, including starvation or treatment stress. Furthermore, long non-coding RNA NEAT1, which has been linked to several stages of carcinogenesis and in all forms of the illness, has drawn attention as a major player in cancer biology. NEAT1 is a structural portion of nuclear paraspeckles and has roles in deactivating expression in both transcriptional and post-transcriptional levels. NEAT1 acts in carcinogenesis in numerous ways, comprising interactions with microRNAs, the influence of gene articulation, regulation of epigenetics, and engagement in signalling cascades. In addition, the complexity of NEAT1's role in cancer occurrence is amplified by its place in regulating cancer stem cells and the tumor microenvironment. NEAT1's interaction with autophagy further complicates the already complicated function of this RNA in cancer biology. NEAT1 has been linked to autophagy in several types of cancer, influencing autophagy pathways and altering its stress response and tumor cell viability. Understanding the interrelation between NEAT1, autophagy, and cancer will enable practitioners to identify novel treatment targets and approaches to disrupt oncogenic processes, reduce the occurrence of treatment resistance, and increase patient survival rates. Specialized treatment strategies and regimens are thus achievable. In the present review, the authors analyze sophisticated relationship schemes in cancer: The NEAT1 pathway and the process of autophagy.
Collapse
Affiliation(s)
- Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia.
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
221
|
Sharma A, Virmani T, Kumar G, Sharma A, Virmani R, Gugulothu D, Singh K, Misra SK, Pathak K, Chitranshi N, Coutinho HDM, Jain D. Mitochondrial signaling pathways and their role in cancer drug resistance. Cell Signal 2024; 122:111329. [PMID: 39098704 DOI: 10.1016/j.cellsig.2024.111329] [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: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Mitochondria, traditionally known as cellular powerhouses, now emerge as critical signaling centers influencing cancer progression and drug resistance. The review highlights the role that apoptotic signaling, DNA mutations, mitochondrial dynamics and metabolism play in the development of resistance mechanisms and the advancement of cancer. Targeted approaches are discussed, with an emphasis on managing mitophagy, fusion, and fission of the mitochondria to make resistant cancer cells more susceptible to traditional treatments. Additionally, metabolic reprogramming can be used to effectively target metabolic enzymes such GLUT1, HKII, PDK, and PKM2 in order to avoid resistance mechanisms. Although there are potential possibilities for therapy, the complex structure of mitochondria and their subtle role in tumor development hamper clinical translation. Novel targeted medicines are put forth, providing fresh insights on combating drug resistance in cancer. The study also emphasizes the significance of glutamine metabolism, mitochondrial respiratory complexes, and apoptotic pathways as potential targets to improve treatment effectiveness against drug-resistant cancers. Combining complementary and nanoparticle-based techniques to target mitochondria has demonstrated encouraging results in the treatment of cancer, opening doors to reduce resistance and enable individualized treatment plans catered to the unique characteristics of each patient. Suggesting innovative approaches such as drug repositioning and mitochondrial drug delivery to enhance the efficacy of mitochondria-targeting therapies, presenting a pathway for advancements in cancer treatment. This thorough investigation is a major step forward in the treatment of cancer and has the potential to influence clinical practice and enhance patient outcomes.
Collapse
Affiliation(s)
- Ashwani Sharma
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Tarun Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India.
| | - Girish Kumar
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India.
| | - Anjali Sharma
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India
| | - Reshu Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India.
| | - Dalapathi Gugulothu
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Shashi Kiran Misra
- School of Pharmaceutical Sciences, CSJM University Kanpur, Kanpur 208024, India
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India
| | - Nitin Chitranshi
- Macquarie Medical School, Macquarie University, New South Wales, Australia; School of Science and Technology, the University of New England, Armidale, New South Wales, Australia.
| | | | - Divya Jain
- Department of Microbiology, School of Applied and Life Sciences, Uttaranchal University, Dehradun 248007, Uttarakhand, India
| |
Collapse
|
222
|
Nasr Esfahani F, Karimi S, Jalilian Z, Alavi M, Aziz B, Alhagh Charkhat Gorgich E, Mozafari MR, Taghavi E, Aminnezhad S, Ataei S. Functionalized and Theranostic Lipidic and Tocosomal Drug Delivery Systems: Potentials and Limitations in Cancer Photodynamic Therapy. Adv Pharm Bull 2024; 14:524-536. [PMID: 39494248 PMCID: PMC11530887 DOI: 10.34172/apb.2024.038] [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: 09/16/2023] [Revised: 02/09/2024] [Accepted: 03/03/2024] [Indexed: 11/05/2024] Open
Abstract
Photodynamic therapy (PDT) is a multidisciplinary area, which involves photophysics and photochemical sciences and plays an important role in cancer diagnosis and treatment. PDT involves a photo-activable drug called photosensitizer (PS), a specific wavelength of light and cellular compounds to produce toxic oxygen species in a much-localized way to destroy malignant tumors. Despite the various benefits of PDT, some PS-related limitations hinder its use as an ideal treatment option for cancer. To address these limitations (e.g., poor bioavailability, weak permeability, hydrophobicity, and aggregation), lipid-based and vesicular drug delivery systems have been employed. These carrier systems possess the ability to enhance the bioavailability, permeability, and solubility of the drug. Furthermore, they tend to load hydrophobic and lipophilic compounds and can be employed for an efficient and targeted drug delivery. The purpose of this review is to highlight the precise idea of PDT, the limitations of PDT related to PS, and the application of lipidic and tocosomal carriers in PDT for the treatment of various types of cancers. Liposomes, nanoliposomes, solid lipid nanoparticles, vesicular phospholipid gels, exosomes, transferosomes, and tocosomes are presented as commonly-employed vesicular drug carriers. Moreover, the amalgamation of cell-based drug delivery systems (CBDDS) with PDT holds considerable potential as an encouraging avenue in cancer treatment, especially in the context of immunotherapy.
Collapse
Affiliation(s)
- Fahime Nasr Esfahani
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Sahand Karimi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan 6617715175, Iran
| | - Zahra Jalilian
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Mehran Alavi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan 6617715175, Iran
| | - Bushra Aziz
- Department of Physics, Women University of Azad Jammu & Kashmir, Bagh 12500, Azad Kashmir, Pakistan
| | - Enam Alhagh Charkhat Gorgich
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - M. R. Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Elham Taghavi
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu (UMT), 21030 Kuala Nerus, Terengganu, Malaysia
| | - Sargol Aminnezhad
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Sara Ataei
- Department of Clinical Pharmacy (Pharmacotherapy), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
223
|
Wosny M, Aeppli S, Fischer S, Peres T, Rothermundt C, Hastings J. Factors Guiding Clinical Decision-Making in Genitourinary Oncology. Cancer Med 2024; 13:e70304. [PMID: 39435678 PMCID: PMC11494402 DOI: 10.1002/cam4.70304] [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: 05/27/2024] [Revised: 07/25/2024] [Accepted: 09/22/2024] [Indexed: 10/23/2024] Open
Abstract
INTRODUCTION Clinical decision-making in oncology is a complex process, with the primary goal of identifying the most effective treatment tailored to individual cancer patients. Many factors influence the treatment decision: disease- and patient-specific criteria, the increasingly complex treatment landscape, market authorization and drug availability, financial aspects, and personal treatment expertise. In the domain of genitourinary cancers, particularly prostate cancer, decision-making is challenging. Despite the prevalence of this malignancy, there are few in-depth explorations of these factors within real-world scenarios. Understanding and refining this intricate decision-making process is essential for future successful clinical decisions and the integration of computerized decision support into clinicians' workflows. AIM The objective of this study is to improve the current knowledge base and evidence of the factors that influence treatment decision-making for patients with genitourinary cancers. METHODS Assessment of how routine treatment decisions are made for genitourinary cancers was performed by a mixed-methods study, encompassing field observations and focus group discussions. RESULTS In total, we identified 59 factors that influence clinical decision-making in oncology, specifically for genitourinary and prostate cancer. Of these, 23 criteria can be classified as decision-maker-related criteria encompassing personal, cognitive, and emotional attributes and factors of both, healthcare professionals and patients. Moreover, 20 decision-specific criteria have been identified that refer to clinical and disease-related factors, followed by 16 contextual decision factors that describe the relevant criteria introduced by the specific circumstances and environment in which the treatment decision is made. CONCLUSION By presenting an exhaustive set of decision factors and providing specific examples for genitourinary cancers, this observational study establishes a possible framework for a better understanding of decision-making. Moreover, we specify and expand the set of decision factors, while emphasizing the importance of cognitive, emotional, and human factors, as well as the quality and accessibility of decision-relevant information.
Collapse
Affiliation(s)
- Marie Wosny
- School of MedicineUniversity of St.Gallen (HSG)St GallenSwitzerland
- Institute for Implementation Science in Health CareUniversity of Zurich (UZH)ZurichSwitzerland
| | - Stefanie Aeppli
- Department of Medical Oncology and HematologyKantonsspital St.Gallen (KSSG)St.GallenSwitzerland
| | - Stefanie Fischer
- Department of Medical Oncology and HematologyKantonsspital St.Gallen (KSSG)St.GallenSwitzerland
| | - Tobias Peres
- Department of Medical Oncology and HematologyKantonsspital St.Gallen (KSSG)St.GallenSwitzerland
| | - Christian Rothermundt
- Department of Medical Oncology and HematologyKantonsspital St.Gallen (KSSG)St.GallenSwitzerland
| | - Janna Hastings
- School of MedicineUniversity of St.Gallen (HSG)St GallenSwitzerland
- Institute for Implementation Science in Health CareUniversity of Zurich (UZH)ZurichSwitzerland
- Swiss Institute of Bioinformatics (SIB)LausanneSwitzerland
| |
Collapse
|
224
|
Kunjalwar R, Keerti A, Chaudhari A, Sahoo K, Meshram S. Microbial Therapeutics in Oncology: A Comprehensive Review of Bacterial Role in Cancer Treatment. Cureus 2024; 16:e70920. [PMID: 39502977 PMCID: PMC11535891 DOI: 10.7759/cureus.70920] [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: 09/26/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
Conventional cancer therapies, including chemotherapy, radiotherapy, and immunotherapy, have significantly advanced cancer treatment. However, these modalities often face limitations such as systemic toxicity, lack of specificity, and the emergence of resistance. Recent advancements in genetic engineering and synthetic biology have rekindled interest in using bacteria as a novel therapeutic approach in oncology. This comprehensive review explores the potential of microbial therapeutics, particularly bacterial therapies, in the treatment of cancer. Bacterial therapies offer several unique advantages, such as the ability to selectively target and colonize hypoxic and necrotic regions of tumors, areas typically resistant to conventional treatments. The review delves into the mechanisms through which bacteria exert antitumor effects, including direct tumor cell lysis, modulation of the immune response, and delivery of therapeutic agents like cytotoxins and enzymes. Various bacterial species, such as Salmonella, Clostridium, Lactobacillus, and Listeria, have shown promise in preclinical and clinical studies, demonstrating diverse mechanisms of action and therapeutic potential. Moreover, the review discusses the challenges associated with bacterial therapies, such as safety concerns, immune evasion, and the need for precise targeting, and how recent advances in genetic engineering are being used to overcome these hurdles. Current clinical trials and combination strategies with conventional therapies are also highlighted to provide a comprehensive overview of the ongoing developments in this field. In conclusion, while bacterial therapeutics present a novel and promising avenue in cancer treatment, further research and clinical validation is required to fully realize their potential. This review aims to inspire further exploration into microbial oncology, paving the way for innovative and more effective cancer therapies.
Collapse
Affiliation(s)
- Radha Kunjalwar
- Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Akshunna Keerti
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Achal Chaudhari
- Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Kaushik Sahoo
- Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Supriya Meshram
- Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
225
|
Tan B, Kartal Y, Yesilyurt F, Akdoğan N, Doyduk D, Dişli A. Synthesis of new phenothiazine derivatives: Molecular docking, assessment of cytotoxic activity and oxidant-antioxidant properties on PCS-201-012, HT-29, and SH-SY5Y cell lines. Arch Pharm (Weinheim) 2024; 357:e2400281. [PMID: 39058899 DOI: 10.1002/ardp.202400281] [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: 04/16/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024]
Abstract
Phenothiazine (PTZ) derivatives have been acknowledged as versatile compounds with significant implications across various areas of medicine, particularly, in cancer research. The cytotoxic effects of synthesized compounds on both normal and cancerous cells, along with their oxidant-antioxidant properties, are pivotal factors in cancer treatment strategies. In the current study, eight new PTZ derivatives were synthesized and the compounds' cytotoxic activities were assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay while the oxidant-antioxidant properties were evaluated by oxidative stress index (OSI) calculation in SH-SY5Y (a human neuroblastoma cell line), HT-29 (a human colorectal adenocarcinoma cell line), and PCS-201-012 (a human primary dermal fibroblast cell line) cells. Consequently, the half-maximal inhibitory concentration (IC50) values of compound 3a were determined to be 218.72, 202.85, and 227.86 μM while the IC50 values of compound 3b were defined to be 227.42, 199.27, and 250.11 μM in PCS-201-012, HT-29, and SH-SY5Y cells, respectively. Additionally, it was determined that the synthesized compounds demonstrated the lowest OSI in PCS-201-012 cells as compared to the other cell lines.
Collapse
Affiliation(s)
- Bensu Tan
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Türkiye
| | - Yasemin Kartal
- Department of Physiology, Faculty of Medicine, Kırklareli University, Kırklareli, Türkiye
| | - Fatma Yesilyurt
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Türkiye
| | - Nurdan Akdoğan
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Türkiye
| | - Doğukan Doyduk
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Türkiye
| | - Ali Dişli
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Türkiye
| |
Collapse
|
226
|
Kim S, Kim S, Kim S, Lee NE, Lee SH, Kim H, Lee H. Improvement of Therapeutic Effect via Inducing Non-Apoptotic Cell Death Using mRNA-Protection Nanocage. Adv Healthc Mater 2024; 13:e2400240. [PMID: 39081097 DOI: 10.1002/adhm.202400240] [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: 01/21/2024] [Revised: 07/22/2024] [Indexed: 10/30/2024]
Abstract
Necroptosis, a cell death mechanism with the characteristics of both apoptosis and necrosis, is proposed as a promising therapeutic approach for cancer therapy. Induction of necroptosis for cancer therapy may be possible through the regulation of the expression of a key factor gene receptor-interacting protein kinase-3 (RIPK3) via in vitro transcription (IVT) mRNA delivery. However, mRNA is susceptible to degradation and has a low delivery efficiency, which highlights the requirement of a proper delivery vehicle for intracellular delivery. Therefore, a new mRNA delivery system based on the nanostructured silica nanoparticles, termed mRNA-protective nanocage (mPN) has been developed. High-efficiency expression of RIPK3 and induction of necroptosis is achieved through delivery of RIPK3 IVT mRNA with mPN in vitro and in vivo models. Importantly, the mPN carrying RIPK3 mRNA distributed locally in tumors upon intravascular injection, and successfully induced necroptosis and immune cell infiltration, a hallmark of necroptosis. the suppression of tumor growth in a murine cancer model, demonstrating the synergistic effect of RIPK3 mRNA- and immune cell-mediated therapy is also observed. These findings suggest the potential for anticancer therapy through necroptosis induction and provide a strategy for the development of mRNA-based nanomedicine.
Collapse
Affiliation(s)
- Seoyoung Kim
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seongchan Kim
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, 52828, Republic of Korea
| | - Sojin Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Nan-Ee Lee
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo-Hwan Lee
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyunkyung Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Hyojin Lee
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- SKKU-KIST, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| |
Collapse
|
227
|
Syed RU, Banu H, Alshammrani A, Alshammari MD, G SK, Kadimpati KK, Khalifa AAS, Aboshouk NAM, Almarir AM, Hussain A, Alahmed FK. MicroRNA-21 (miR-21) in breast cancer: From apoptosis dysregulation to therapeutic opportunities. Pathol Res Pract 2024; 262:155572. [PMID: 39226804 DOI: 10.1016/j.prp.2024.155572] [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: 07/31/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Breast cancer, a pervasive and complex disease, continues to pose significant challenges in the field of oncology. Its heterogeneous nature and diverse molecular profiles necessitate a nuanced understanding of the underlying mechanisms driving tumorigenesis and progression. MicroRNA-21 (miR-21) has emerged as a crucial player in breast cancer development and progression by modulating apoptosis, a programmed cell death mechanism that eliminates aberrant cells. MiR-21 overexpression is a hallmark of breast cancer, and it is associated with poor prognosis and resistance to conventional therapies. This miRNA exerts its oncogenic effects by targeting various pro-apoptotic genes, including Fas ligand (FasL), programmed cell death protein 4 (PDCD4), and phosphatase and tensin homolog (PTEN). By suppressing these genes, miR-21 promotes breast cancer cell survival, proliferation, invasion, and metastasis. The identification of miR-21 as a critical regulator of apoptosis in breast cancer has opened new avenues for therapeutic intervention. This review investigates the intricate mechanisms through which miR-21 influences apoptosis, offering insights into the molecular pathways and signaling cascades involved. The dysregulation of apoptosis is a hallmark of cancer, and understanding the role of miR-21 in this context holds immense therapeutic potential. Additionally, the review highlights the clinical significance of miR-21 as a diagnostic and prognostic biomarker in breast cancer, underscoring its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - Humera Banu
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia.
| | - Alia Alshammrani
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Satheesh Kumar G
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Kishore Kumar Kadimpati
- Department of Environmental Biotechnology, Faculty of Energy and Environmental Engineering, The Silesian University of Technology, Poland
| | - Amna Abakar Suleiman Khalifa
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | - Arshad Hussain
- Department of Clinical Pharmacy, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia
| | - Farah Khaled Alahmed
- Department of Clinical Pharmacy, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia
| |
Collapse
|
228
|
Rodrigues P, Bangali H, Ali E, Sharma MK, Abdullaev B, Alkhafaji AT, Deorari MM, Zabibah RS, Haslany A. Microproteins/micropeptides dysregulation contributes to cancer progression and development: A mechanistic review. Cell Biol Int 2024; 48:1395-1405. [PMID: 39010637 DOI: 10.1002/cbin.12219] [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: 10/04/2023] [Revised: 05/06/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
Microproteins, known as micropeptides, are small protein molecules encoded by short open reading frames. These recently identified molecules have been proven to be an essential part of the human proteome that participates in multiple processes, such as DNA repair, mitochondrial respiration, and regulating different signaling pathways. A growing body of studies has evidenced that microproteins exhibit dysregulated expression levels in various malignancies and contribute to tumor progression. It has been reported that microproteins interact with many proteins, such as enzymes (e.g., adenosine triphosphate synthase) and signal transducers (e.g., c-Jun), and regulate malignant cell metabolism, proliferation, and metastasis. Moreover, microproteins have been found to play a significant role in multidrug resistance in vitro and in vivo by their activity in DNA repair pathways. Considering that, this review intended to summarize the roles of microproteins in different aspects of tumorigenesis with diagnostic and therapeutic perspectives.
Collapse
Affiliation(s)
- Paul Rodrigues
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Harun Bangali
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Saudi Arabia
| | - Eyhab Ali
- College of Chemistry, Al-Zahraa University for Women, Karbala, Iraq
| | - M K Sharma
- Chaudhary Charan Singh University, Meerut, Uttar Pradesh, India
| | - Bekhzod Abdullaev
- Department of Biotechnology, New Uzbekistan University, Tashkent, Uzbekistan
| | | | - Maha Medha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Rahman S Zabibah
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Haslany
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| |
Collapse
|
229
|
Kim J, Jia X. Flexible multimaterial fibers in modern biomedical applications. Natl Sci Rev 2024; 11:nwae333. [PMID: 39411353 PMCID: PMC11476783 DOI: 10.1093/nsr/nwae333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/19/2024] Open
Abstract
Biomedical devices are indispensable in modern healthcare, significantly enhancing patients' quality of life. Recently, there has been a drastic increase in innovations for the fabrication of biomedical devices. Amongst these fabrication methods, the thermal drawing process has emerged as a versatile and scalable process for the development of advanced biomedical devices. By thermally drawing a macroscopic preform, which is meticulously designed and integrated with functional materials, hundreds of meters of multifunctional fibers are produced. These scalable flexible multifunctional fibers are embedded with functionalities such as electrochemical sensing, drug delivery, light delivery, temperature sensing, chemical sensing, pressure sensing, etc. In this review, we summarize the fabrication method of thermally drawn multifunctional fibers and highlight recent developments in thermally drawn fibers for modern biomedical application, including neural interfacing, chemical sensing, tissue engineering, cancer treatment, soft robotics and smart wearables. Finally, we discuss the existing challenges and future directions of this rapidly growing field.
Collapse
Affiliation(s)
- Jongwoon Kim
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24060, USA
| | - Xiaoting Jia
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24060, USA
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24060, USA
- Department of Materials Science and Engineering, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
230
|
Sahin C, Mutlu D, Erdem A, Kilincarslan R, Arslan S. New cyclometalated iridium(III) complexes bearing substituted 2-(1H-benzimidazol-2-yl)quinoline: Synthesis, characterization, electrochemical and anticancer studies. Bioorg Chem 2024; 151:107706. [PMID: 39128244 DOI: 10.1016/j.bioorg.2024.107706] [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: 06/28/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
New iridium(III) compounds (C1-C3) bearing 2-(1H-benzimidazol-2-yl)quinoline ligands with different side groups (benzyl, 2,3,4,5,6-pentamethylbenzyl and 2,3,4,5,6-pentafluorobenzyl) were synthesized and characterized by using spectroscopic analyses. The effects of different side groups of iridium compounds on the photophysical and electrochemical properties have been investigated. The cytotoxicity and apoptosis of the compounds have been evaluated on breast cancer cell lines using various methods including MTT assay, flow cytometry, qRT-PCR, and colony formation. The cytotoxicity of C1, expressed as IC50 values, was found to be 11.76 μM for MDA-MB-231 and 5.35 μM for MCF-7 cells. For C3, the IC50 value was 16.22 μM for MDA-MB-231 and 8.85 μM for MCF-7 cells. In both cell lines, increased levels of Bax and caspase 3, along with downregulation of BCL-2 and positive annexin V staining, were observed, confirming apoptosis. Moreover, the colony-forming abilities in both cell lines decreased after C1 and C3 complex treatment. All these results suggest that the compounds C1 and C3 may have potential in the treatment of breast cancer, though further research is needed to confirm their efficacy.
Collapse
Affiliation(s)
- Cigdem Sahin
- Department of Engineering Basic Sciences, Faculty of Engineering and Natural Sciences, Istanbul Medeniyet University, Istanbul, Turkey.
| | - Dogukan Mutlu
- Department of Biology, Faculty of Science, Pamukkale University, Denizli, Turkey.
| | - Ahmet Erdem
- Advanced Technology Application and Research Center, Pamukkale University, Denizli, Turkey
| | - Rafet Kilincarslan
- Department of Chemistry, Faculty of Science, Pamukkale University, Denizli, Turkey.
| | - Sevki Arslan
- Department of Biology, Faculty of Science, Pamukkale University, Denizli, Turkey
| |
Collapse
|
231
|
Li S, Amakye WK, Zhao Z, Xin X, Jia Y, Zhang H, Ren Y, Zhou Y, Zhai L, Kang W, Lu X, Guo J, Wang M, Xu Y, Yi J, Ren J. Prognostic value of anthropometric- and biochemistry-based nutrition status indices on blood chemistry panel levels during cancer treatment. Nutrition 2024; 126:112520. [PMID: 39111096 DOI: 10.1016/j.nut.2024.112520] [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: 01/22/2024] [Revised: 05/24/2024] [Accepted: 06/08/2024] [Indexed: 09/10/2024]
Abstract
Body weight, body mass index (BMI), Nutrition Risk Screening 2002 (NRS2002), and prognostic nutritional index (PNI) are among vital nutrition status indices employed during cancer treatment. These have also been associated with levels of blood chemistry panels (BCPs), which are touted as significant indicators of disease prognosis. However, it remains unclear which nutrition status index better predicts future trends in specific BCPs. Using the records of 407 cancer patients, we retrospectively examined the potential of nutritional status indices at baseline for predicting changes in specific BCPs over a 6-week period. Generally, both serum biochemical parameters and nutrition status indices fluctuated over the study period among study participants. PNI was often linearly associated with blood cell counts (white blood cells [WBCs] and hemoglobin) compared with anthropometric-based nutrition status indices. Increase in body weight was protective against having abnormal lymphocyte levels at 6 weeks (odds ratio [OR]: 0.960-0.974; CI: 0.935-0.997; P < 0.05), while increase in baseline PNI was associated with 0.865-0.941 and 0.675-0.915 odds of having future abnormal WBC and lymphocyte levels, respectively. Increases in PNI were also protective against having future abnormal albumin levels (OR: 0.734-0.886) and 8.5-12.5% decreases in the odds of having an abnormal C-reactive protein level in subsequent visits. Changes in NRS2002 tended to be associated with the odds of having future abnormal blood glucose levels. In conclusion, the serum biochemistry-derived nutrition status index, PNI, is a more consistent measure as an early indicator to track the trends of future changes in the BCPs of cancer patients. This implies that PNI could be targeted as an early-warning measure with relevant preventive interventions for patients at risk of malnutrition.
Collapse
Affiliation(s)
- Suyun Li
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Beijing, 100021, China
| | - William Kwame Amakye
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Zikuan Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Xiaowei Xin
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Beijing, 100021, China
| | - Ying Jia
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Beijing, 100021, China
| | - Hui Zhang
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Beijing, 100021, China
| | - Yuwei Ren
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Langfang, 065001, China
| | - Yuxin Zhou
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Langfang, 065001, China
| | - Lina Zhai
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Langfang, 065001, China
| | - Weicong Kang
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Langfang, 065001, China
| | - Xuemin Lu
- Department of Clinical Nutrition, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Langfang, 065001, China
| | - Ji Guo
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Min Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Yongzhao Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Junlin Yi
- Department of Radiotherapy, National Cancer Hospital/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Science, Beijing, 100021, China
| | - Jiaoyan Ren
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China.
| |
Collapse
|
232
|
Ghoshal A, Prince R, Downar J, Lapenskie J, Subramaniam S, Wegier P, Le L, Hannon B. Exploring the Utility of the Modified Hospitalized-Patient One-Year Mortality Risk Score to Trigger Referrals to Palliative Care for Inpatients With Cancer. Cancer Med 2024; 13:e70292. [PMID: 39382260 PMCID: PMC11462593 DOI: 10.1002/cam4.70292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Estimating prognosis can be a barrier to timely palliative care involvement. The modified Hospitalized-patient One-year Mortality Risk (mHOMR) score uses hospital admission data to calculate the risk of death within 12 months and may be a useful tool to trigger a referral to palliative care. METHODS The mHOMR tool was retrospectively applied to consecutive acute admissions to a quaternary cancer center in Toronto, Canada from March 1 to May 31, 2018. The study aimed to investigate the association between dichotomized mHOMR scores (the cohort median score of 0.27 and the developer-recommended score of 0.21) and the risk of death, and whether these could be used to identify patients who may benefit from timely palliative care involvement. RESULTS Of 269 inpatients, 87 were elective admissions and excluded from further analyses. At the median mHOMR score of 0.27, 91/182 patients (50%) were categorized as high-risk of death within 12 months (mHOMR+), 53 (58%) were referred to palliative care. At the lower cut-off of 0.21, 103 patients were mHOMR+, of whom 57 (55.3%) were referred to palliative care. The higher mHOMR was significantly associated with mortality (29.7% mHOMR- vs. 39.8% mHOMR+ at 12 months, log-rank p < 0.05). The association between the developer-recommended mHOMR cut-off (≥ 0.21) and mortality was not significant (p = 0.15). CONCLUSIONS A higher mHOMR score was significantly associated with the risk of mortality in patients with advanced cancer. However, the developer-recommended mHOMR cut-off of 0.21 failed to identify a statistically significant difference between patients with advanced cancer at low versus high scores. While mHOMR may be a useful tool to augment clinical judgment and identify inpatients with advanced cancer at high risk of death, who in turn may benefit from referral to palliative care, the optimal mHOMR cutoff may warrant adjustment for this population.
Collapse
Affiliation(s)
- A. Ghoshal
- Princess Margaret Cancer CentreTorontoOntarioCanada
- Department of Medicine, Division of Palliative MedicineUniversity of TorontoTorontoOntarioCanada
| | - R. Prince
- Epworth Cancer Services Clinical InstituteGeelongVictoriaAustralia
| | - J. Downar
- Department of Medicine, Division of Palliative CareUniversity of OttawaOttawaOntarioCanada
- Ottawa Hospital Research Institute and Bruyère Research InstituteOttawaOntarioCanada
| | - J. Lapenskie
- Ottawa Hospital Research Institute and Bruyère Research InstituteOttawaOntarioCanada
| | | | - P. Wegier
- Humber River HealthNorth YorkOntarioCanada
| | - L. W. Le
- Department of BiostatisticsPrincess Margaret Cancer CentreTorontoOntarioCanada
| | - B. Hannon
- Princess Margaret Cancer CentreTorontoOntarioCanada
- Department of Medicine, Division of Palliative MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
233
|
Mustafa EM, Shahin AI, Alrashed AS, Bahaaddin AH, Alajmi AA, Hashem O, Anbar HS, El-Gamal MI. An overview of the latest outlook of sulfamate derivatives as anticancer candidates (2020-2024). Arch Pharm (Weinheim) 2024; 357:e2400331. [PMID: 38943437 DOI: 10.1002/ardp.202400331] [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: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 07/01/2024]
Abstract
Considering the emergence of new anticancer drugs, in this review we emphasized and highlighted the recent reports and advances related to sulfamate-incorporating compounds with potential anticancer activity during the last 5 years (2020-2024). Additionally, we discussed their structure-activity relationship, clarifying their potent bioactivity as anticancer agents. Sulfamate derivatives hold promise as effective therapeutic candidates against cancer. By targeting biological targets associated with the development of cancer, such as steroid sulfatases (STS), carbonic anhydrases (CAs), microtubules, NEDD8-activating enzyme, small ubiquitin-like modifiers (SUMO)-activating enzyme (SAE), cyclin-dependent kinases (CDKs), breast cancer susceptibility gene 1 (BRCA1), and so on, this can furnish small molecules as anticancer lead candidates serving the drug discovery field. For example, compound 2, an STS inhibitor, demonstrated superior activity compared to its reference, irosustat, by fivefold. In addition, compound 21, an SAE, is under phase I clinical trials. Continued research into sulfamate derivatives holds potential for the development of novel therapeutic agents targeting various diseases.
Collapse
Affiliation(s)
- Esra M Mustafa
- Research, Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Afnan I Shahin
- Research, Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Aishah S Alrashed
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Aesheh H Bahaaddin
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Aljawhra A Alajmi
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Omar Hashem
- Research, Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hanan S Anbar
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Mohammed I El-Gamal
- Research, Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
234
|
Kangari P, Salahlou R, Vandghanooni S. Harnessing the Therapeutic Potential of Mesenchymal Stem Cells in Cancer Treatment. Adv Pharm Bull 2024; 14:574-590. [PMID: 39494266 PMCID: PMC11530882 DOI: 10.34172/apb.2024.052] [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] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 11/05/2024] Open
Abstract
Cancer, as a complicated disease, is considered to be one of the major leading causes of death globally. Although various cancer therapeutic strategies have been established, however, some issues confine the efficacies of the treatments. In recent decades researchers for finding efficient therapeutic solutions have extensively focused on the abilities of stem cells in cancer inhibition. Mesenchymal stem cells (MSCs) are multipotent stromal cells that can the most widely extracted from various sources such as the bone marrow (BM), placenta, umbilical cord (UC), menses blood, Wharton's jelly (WJ), adipose tissue and dental pulp (DP). These cells are capable of differentiating into the osteoblasts, chondrocytes, and adipocytes. Due to the unique characteristics of MSCs such as paracrine effects, immunomodulation, tumor-tropism, and migration, they are considered promising candidates for cancer therapeutics. Currently, MSCs are an excellent living carrier for delivery of therapeutic genes and chemical agents to target tumor sites. Also, exosomes, the most important extracellular vesicle released from MSCs, act as a strong cell-free tool for cancer therapeutics. MSCs can prevent cancer progression by inhibiting several signaling pathways, such as wnt/β-catenin and PI3K/AKT/mTOR. However, there are several challenges associated with the use of MSCs and their exosomes in the field of therapy that need to be considered. This review explores the significance of MSCs in cell-based therapy, focusing on their homing properties and immunomodulatory characteristics. It also examines the potential of using MSCs as carriers for delivery of anticancer agents and their role in modulating the signal transduction pathways of cancer cells.
Collapse
Affiliation(s)
- Parisa Kangari
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Salahlou
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
235
|
Kwon JA, Yang S, Koh SJ, Noh YJ, Kang DY, Yang SB, Kwon EJ, Seo JW, Kim JS, Ock M. Development and Feasibility Evaluation of Smart Cancer Care 2.0 Based on Patient-Reported Outcomes for Post-Discharge Management of Patients with Cancer. Cancer Res Treat 2024; 56:1040-1049. [PMID: 38605663 PMCID: PMC11491237 DOI: 10.4143/crt.2024.003] [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: 01/02/2024] [Accepted: 04/07/2024] [Indexed: 04/13/2024] Open
Abstract
PURPOSE A "Smart Cancer Care" platform that integrates patient-reported outcomes (PROs) with management has been established in Korea. This study focused on improving health behaviors and connecting patients to welfare services by introducing and assessing the feasibility of "Smart Cancer Care 2.0," an enhanced version designed for monitoring complications post-cancer treatment. MATERIALS AND METHODS Smart Cancer Care 2.0 was developed by conducting a literature review and consulting with expert panels to identify symptoms or variables requiring monitoring and management guidelines based on the treatment type. Qualitative and quantitative surveys were conducted to assess the feasibility of the app and web system based on the experiences of patients with cancer and healthcare workers. RESULTS A total of 81 symptoms or variables (chemotherapy-, surgery-, radiotherapy-, rehabilitation-, and health management-related) were selected for management in Smart Cancer Care 2.0. PROs for these symptoms were basically categorized into three severity grades: preventive management, self-treatment, and consultation with a healthcare worker or visit to a healthcare institution. The overall mean scores in the feasibility evaluation by patients and healthcare workers were 3.83 and 3.90 points, respectively, indicating high usefulness. CONCLUSION Smart Cancer Care 2.0 leverages the existing information and communication technologies-based platform, Smart Cancer Care, and further includes health behaviors and welfare services. Smart Cancer Care 2.0 may play a crucial role in establishing a comprehensive post-discharge management system for patients with cancer as it provides suitable interventions based on patients' responses and allows the regularly collected PROs to be easily viewed for streamlined care.
Collapse
Affiliation(s)
- Jin Ah Kwon
- Department of Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Songsoo Yang
- Department of Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Su-Jin Koh
- Division of Hematology and Oncology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Young Ju Noh
- Department of Radiation Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Dong Yoon Kang
- Department of Preventive Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
- Department of Preventive Medicine, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, Korea
| | - Sol Bin Yang
- Department of Preventive Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Eun Ji Kwon
- Department of Preventive Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Jeong-Wook Seo
- Public Health and Medical Services Team, Ulsan University Hospital, Ulsan, Korea
| | - Jin Sung Kim
- Department of Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Minsu Ock
- Department of Preventive Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
- Department of Preventive Medicine, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
236
|
Rawas-Qalaji M, Jagal J, Sadik S, Said Z, Ahmed IS, Haider M, Hussain Z, Alhalaweh A. Assessment of enhancing curcumin's solubility versus uptake on its anti-cancer efficacy. Colloids Surf B Biointerfaces 2024; 242:114090. [PMID: 39018909 DOI: 10.1016/j.colsurfb.2024.114090] [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: 05/12/2024] [Revised: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Curcumin (CUR) exhibits anti-inflammatory and anti-cancer activities. However, its poor solubility and bioavailability limit its therapeutic applications. Several CUR nano-formulations have been developed to enhance its solubility and uptake, thereby improving its anti-cancer activity. Despite this, studies comparing the effect of enhanced CUR solubility versus cellular uptake on its anti-cancer efficacy are lacking. Therefore, CUR nanofibers (CUR NF) were synthesized by electrospinning using a water-soluble polymer to enhance CUR solubility. While CUR nanoparticles (CUR NP) were synthesized by nanoprecipitation method using a water-insoluble polymer to enhance CUR cellular uptake. Both nano-formulations aim to improve CUR cellular concentration and anti-cancer activity against various cancer cells. CUR NF and CUR NP were successfully synthesized at drug load (DL%) of 10 %, 20 %, and 40 % w/w. Both nano-formulations were characterized, and CUR dissolution, release, cytotoxicity, IC50, and cellular uptake were assessed. A gradual increase in NF diameter and NP size was observed as the drug load% increased compared to the placebo. NF showed a rapid CUR release and increased solubility by 16-38 fold. In contrast, NP sustained CUR release and resulted in only a 2-fold increase in solubility. Both formulations significantly reduced cell viability and IC50 compared to free CUR. However, CUR NP demonstrated higher cell toxicity (70-80 %) than CUR NF (60 %) and reduced IC50 up to 4 μM compared to 11 μM for NF. Enhancing CUR solubility or uptake can significantly increase its cellular concentration and anti-cancer activity. However, enhancing CUR cellular uptake by NP demonstrated superior anti-cancer effect compared to enhancing its solubility by NF.
Collapse
Affiliation(s)
- Mutasem Rawas-Qalaji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates; Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA.
| | - Jayalakshmi Jagal
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Sefeera Sadik
- Research Institute of Science and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Zafar Said
- Research Institute of Science and Engineering, University of Sharjah, Sharjah, United Arab Emirates; Department of Sustainable & Renewable Energy Engineering, University of Sharjah, Sharjah, United Arab Emirates; Department of Industrial and Mechanical Engineering, Lebanese American University (LAU), Byblos, Lebanon
| | - Iman Saad Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohamed Haider
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Zahid Hussain
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Amjad Alhalaweh
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates; Department of Pharmacy, Uppsala University, Biomedical Centre, Uppsala SE-751 23, Sweden
| |
Collapse
|
237
|
Haroon M, Kang SC. Kaempferol Synergistically Enhances Cisplatin-induced Apoptosis and Cell Cycle Arrest in Colon Cancer Cells. J Cancer Prev 2024; 29:69-87. [PMID: 39398110 PMCID: PMC11467758 DOI: 10.15430/jcp.24.013] [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: 07/26/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Colon cancer remains a significant global health concern, necessitating the continuous exploration of novel therapeutic strategies. Cisplatin is a first-line chemotherapy medication that is frequently used to treat patients for a variety of malignancies, including colon cancer. However, a major obstacle to its clinical usefulness is acquired resistance. This research investigates the synergistic effects of kaempferol, a natural flavonoid with known anti-cancer properties, in combination with cisplatin, in colon cancer cells. Our study employed colon cancer cell lines to evaluate the individual and combined cytotoxic effects of kaempferol and cisplatin. The results demonstrated a notable enhancement in the cytotoxicity of colon cancer cells when treated with a combination of kaempferol and cisplatin compared to individual treatments. This synergistic effect was further characterized by an increase in apoptosis, as evidenced by morphological changes and biochemical markers of apoptosis and cell cycle. The investigations revealed that the combined treatment led to the modulation of key apoptotic pathways, including the upregulation of pro-apoptotic factors and downregulation of anti-apoptotic factors. Additionally, the synergistic effect was associated with the inhibition of cell proliferation and induction of cell cycle arrest. The findings of this study suggest that the combination of kaempferol and cisplatin holds promise as a potent therapeutic strategy for colon cancer treatment, potentially enhancing the efficacy of conventional chemotherapy while minimizing adverse effects. Further in-depth investigations, including in vivo studies, are warranted to validate these findings and explore the translational potential of this synergistic approach in clinical settings.
Collapse
Affiliation(s)
- Muhammad Haroon
- Department of Biotechnology, Daegu University, Gyeongsan, Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Korea
| |
Collapse
|
238
|
Osorio HM, Castillo-Solís F, Barragán SY, Rodríguez-Pólit C, Gonzalez-Pastor R. Graphene Quantum Dots from Natural Carbon Sources for Drug and Gene Delivery in Cancer Treatment. Int J Mol Sci 2024; 25:10539. [PMID: 39408866 PMCID: PMC11476599 DOI: 10.3390/ijms251910539] [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: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer therapy is constantly evolving, with a growing emphasis on targeted and efficient treatment options. In this context, graphene quantum dots (GQDs) have emerged as promising agents for precise drug and gene delivery due to their unique attributes, such as high surface area, photoluminescence, up-conversion photoluminescence, and biocompatibility. GQDs can damage cancer cells and exhibit intrinsic photothermal conversion and singlet oxygen generation efficiency under specific light irradiation, enhancing their effectiveness. They serve as direct therapeutic agents and versatile drug delivery platforms capable of being easily functionalized with various targeting molecules and therapeutic agents. However, challenges such as achieving uniform size and morphology, precise bandgap engineering, and scalability, along with minimizing cytotoxicity and the environmental impact of their production, must be addressed. Additionally, there is a need for a more comprehensive understanding of cellular mechanisms and drug release processes, as well as improved purification methods. Integrating GQDs into existing drug delivery systems enhances the efficacy of traditional treatments, offering more efficient and less invasive options for cancer patients. This review highlights the transformative potential of GQDs in cancer therapy while acknowledging the challenges that researchers must overcome for broader application.
Collapse
Affiliation(s)
- Henrry M. Osorio
- Departamento de Física, Escuela Politécnica Nacional, Av. Ladrón de Guevara E11-253, Quito 170525, Ecuador; (H.M.O.); (S.Y.B.)
| | - Fabián Castillo-Solís
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (F.C.-S.); (C.R.-P.)
| | - Selena Y. Barragán
- Departamento de Física, Escuela Politécnica Nacional, Av. Ladrón de Guevara E11-253, Quito 170525, Ecuador; (H.M.O.); (S.Y.B.)
| | - Cristina Rodríguez-Pólit
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (F.C.-S.); (C.R.-P.)
- Escuela de Salud Pública, Universidad San Francisco de Quito USFQ, Quito 170527, Ecuador
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito 170403, Ecuador
| | - Rebeca Gonzalez-Pastor
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (F.C.-S.); (C.R.-P.)
| |
Collapse
|
239
|
Alqarni A, Hosmani J, Alassiri S, Alqahtani AMA, Assiri HA. A Network Pharmacology Identified Metastasis Target for Oral Squamous Cell Carcinoma Originating from Breast Cancer with a Potential Inhibitor from F. sargassaceae. Pharmaceuticals (Basel) 2024; 17:1309. [PMID: 39458948 PMCID: PMC11510435 DOI: 10.3390/ph17101309] [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: 08/13/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
This study aimed to identify specific therapeutic targets for oral squamous cell carcinoma (OSCC) that metastasize from breast cancer (BC) by using network pharmacology. The Gene Expression Omnibus for OSCC and BC served as the source of gene expression datasets and their analysis. Upregulated genes and the common intersecting genes of these cancers were determined along with that of the phytochemicals of F. sargassum to predict the pharmacological targets. Further, gene enrichment analysis revealed that their metastasis signature and metastasis targets were determined via a protein interaction network. Molecular docking and pharmacokinetic screening determined the potential therapeutic phytochemicals against the targets. The interaction network of 39 genes thus identified encoding proteins revealed HIF1A as a prominent metastasis target due to its high degree of connectivity and its involvement in cancer-related pathways. Molecular docking showed a strong binding affinity of isonahocol D2, a sargassum-derived compound with HIF1A, presenting a binding energy of -7.1 kcal/mol. Further, pharmacokinetic screening showed favorable ADME properties and molecular dynamics simulations showed stable interactions between isonahocol D2 and HIF1A, with significant stability over 100 ns. This study's results emphasized that isonahocol D2 is a promising therapeutic candidate against HIF1A in OSCC metastasized from breast cancer in translational medicine.
Collapse
Affiliation(s)
| | - Jagadish Hosmani
- Department of Diagnostic Dental Sciences & Oral Biology, College of Dentistry, King Khalid University, Abha 61421, Saudi Arabia; (A.A.); (S.A.); (A.M.A.A.); (H.A.A.)
| | | | | | | |
Collapse
|
240
|
Dabbousy R, Rima M, Roufayel R, Rahal M, Legros C, Sabatier JM, Fajloun Z. Plant Metabolomics: The Future of Anticancer Drug Discovery. Pharmaceuticals (Basel) 2024; 17:1307. [PMID: 39458949 PMCID: PMC11510165 DOI: 10.3390/ph17101307] [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: 07/10/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Drug development from medicinal plants constitutes an important strategy for finding natural anticancer therapies. While several plant secondary metabolites with potential antitumor activities have been identified, well-defined mechanisms of action remained uncovered. In fact, studies of medicinal plants have often focused on the genome, transcriptome, and proteome, dismissing the relevance of the metabolome for discovering effective plant-based drugs. Metabolomics has gained huge interest in cancer research as it facilitates the identification of potential anticancer metabolites and uncovers the metabolomic alterations that occur in cancer cells in response to treatment. This holds great promise for investigating the mode of action of target metabolites. Although metabolomics has made significant contributions to drug discovery, research in this area is still ongoing. In this review, we emphasize the significance of plant metabolomics in anticancer research, which continues to be a potential technique for the development of anticancer drugs in spite of all the challenges encountered. As well, we provide insights into the essential elements required for performing effective metabolomics analyses.
Collapse
Affiliation(s)
- Ranin Dabbousy
- Laboratory of Applied Biotechnology (LBA3B), Department of Cell Culture, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon;
| | - Mohamad Rima
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon;
| | - Rabih Roufayel
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| | - Mohamad Rahal
- School of Pharmacy, Lebanese International University, Beirut 146404, Lebanon;
| | - Christian Legros
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Faculty of Medicine, University Angers, 49000 Angers, France;
| | - Jean-Marc Sabatier
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Université, 13385 Marseille, France
| | - Ziad Fajloun
- Laboratory of Applied Biotechnology (LBA3B), Department of Cell Culture, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon;
- Department of Biology, Faculty of Sciences 3, Campus Michel Slayman Ras Maska, Lebanese University, Tripoli 1352, Lebanon
| |
Collapse
|
241
|
Shams A. Leveraging State-of-the-Art AI Algorithms in Personalized Oncology: From Transcriptomics to Treatment. Diagnostics (Basel) 2024; 14:2174. [PMID: 39410578 PMCID: PMC11476216 DOI: 10.3390/diagnostics14192174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Continuous breakthroughs in computational algorithms have positioned AI-based models as some of the most sophisticated technologies in the healthcare system. AI shows dynamic contributions in advancing various medical fields involving data interpretation and monitoring, imaging screening and diagnosis, and treatment response and survival prediction. Despite advances in clinical oncology, more effort must be employed to tailor therapeutic plans based on each patient's unique transcriptomic profile within the precision/personalized oncology frame. Furthermore, the standard analysis method is not compatible with the comprehensive deciphering of significant data streams, thus precluding the prediction of accurate treatment options. METHODOLOGY We proposed a novel approach that includes obtaining different tumour tissues and preparing RNA samples for comprehensive transcriptomic interpretation using specifically trained, programmed, and optimized AI-based models for extracting large data volumes, refining, and analyzing them. Next, the transcriptomic results will be scanned against an expansive drug library to predict the response of each target to the tested drugs. The obtained target-drug combination/s will be then validated using in vitro and in vivo experimental models. Finally, the best treatment combination option/s will be introduced to the patient. We also provided a comprehensive review discussing AI models' recent innovations and implementations to aid in molecular diagnosis and treatment planning. RESULTS The expected transcriptomic analysis generated by the AI-based algorithms will provide an inclusive genomic profile for each patient, containing statistical and bioinformatics analyses, identification of the dysregulated pathways, detection of the targeted genes, and recognition of molecular biomarkers. Subjecting these results to the prediction and pairing AI-based processes will result in statistical graphs presenting each target's likely response rate to various treatment options. Different in vitro and in vivo investigations will further validate the selection of the target drug/s pairs. CONCLUSIONS Leveraging AI models will provide more rigorous manipulation of large-scale datasets on specific cancer care paths. Such a strategy would shape treatment according to each patient's demand, thus fortifying the avenue of personalized/precision medicine. Undoubtedly, this will assist in improving the oncology domain and alleviate the burden of clinicians in the coming decade.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; or ; Tel.: +00966-548638099
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
242
|
Mierczak K, Garus-Pakowska A. An Overview of Apple Varieties and the Importance of Apple Consumption in the Prevention of Non-Communicable Diseases-A Narrative Review. Nutrients 2024; 16:3307. [PMID: 39408274 PMCID: PMC11478947 DOI: 10.3390/nu16193307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Non-communicable diseases such as cardiovascular diseases, cancers, diabetes, and asthma are increasingly common due to factors like industrialization, urbanization, fast-paced life, stress, sedentary lifestyle, and unbalanced diet in the 21st century. These chronic conditions are a global epidemic, being among the top causes of death worldwide. Preventing these diseases through a nutritious diet is crucial, and scientific studies suggest that appropriate fruit intake, particularly apples, can lower the risk of various health issues. Apples, rich in bioactive compounds, vitamins, minerals, and dietary fiber, offer numerous health benefits. Regular consumption of apples helps reduce the risk of atherosclerosis, coronary artery disease, heart attacks, and diabetes, and also provides anti-asthmatic and anti-allergic effects. Apples aid in detoxification, improve digestion, enhance skin, hair, and nail health, and offer protection against cancers, Alzheimer's, and Parkinson's disease. Apples have been a dietary staple for centuries, consumed in various forms like juices, sauces, and ciders. The reviewed article emphasizes the health benefits of apples, highlighting their role in preventing civilization diseases. It also discusses the characteristics of common apple varieties and the impact of thermal processing on their nutritional content.
Collapse
Affiliation(s)
| | - Anna Garus-Pakowska
- Department of Nutrition and Epidemiology, Medical University of Lodz, 90-752 Lodz, Poland;
| |
Collapse
|
243
|
Zang Y, Qiu Y, Sun Y, Fan Y. Immunomodulatory effects of Huaier granule in cancer therapy: a meta-analysis of randomized controlled trials. Eur J Med Res 2024; 29:467. [PMID: 39342351 PMCID: PMC11438288 DOI: 10.1186/s40001-024-02060-7] [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: 01/10/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND This meta-analysis aimed to summarize the immunomodulatory effect of Huaier (Trametes robiniophila Murr) granule as adjuvant therapy in patients with cancer. MATERIALS AND METHODS: Two authors conducted a search for literature indexed on various databases including PubMed, Embase, Cochrane Library, CNKI, Sinomed, VIP, and WanFang. Randomized controlled trials (RCTs) that investigated the immunomodulatory effect of Huaier granule as adjuvant therapy in cancer patients were included. The outcome of interest included T-lymphocyte subsets (CD3+, CD4+, CD8+ and CD4+/CD8+), immunoglobulin (IgA, IgG, IgM), and natural killer (NK) cells. RESULTS We identified 29 RCTs involving a total of 2206 cancer (including hepatocellular, breast, gastric, colorectal, lung, or nasopharyngeal carcinoma) patients. Compared with conventional treatment alone, Huaier combined conventional treatment significantly improved CD3+ (mean difference [MD] 6.95; 95% confidence intervals [CI] 4.42-9.48), CD4+ (MD 5.53; 95%CI 4.22-6.83), CD4+/CD8+ (MD 0.35; 95%CI 0.25-0.45), IgA (standardized mean difference [SMD] 1.18; 95%CI 0.44-1.93), IgG(SMD 1.71; 95%CI 1.11-2.30), IgM (SMD 0.83; 95%CI 0.59-1.07), and NK cells (MD 5.01; 95%CI 3.61-6.40). However, the effect of Huaier on CD8+ (MD - 1.35; 95%CI - 2.80 to 0.11) was not statistically significant between the groups. CONCLUSIONS Huaier granule as adjuvant therapy may significantly improve immune function in patients with cancers. However, additional well-designed RCTs are needed to validate the current findings considering the methodological flaws of the analyzed trials.
Collapse
Affiliation(s)
- Ye Zang
- Department of Oncology, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, 212399, China
| | - Yue Qiu
- Institute of Molecular Biology and Translational Medicine, Cancer Institute, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, China
| | - Yimeng Sun
- Institute of Molecular Biology and Translational Medicine, Cancer Institute, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, China.
| | - Yu Fan
- Institute of Molecular Biology and Translational Medicine, Cancer Institute, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, China.
| |
Collapse
|
244
|
Chrabąszcz K, Pogoda K, Cieżak K, Panek A, Kwiatek WM. Sensing Biomolecules Associated with Cells' Radiosusceptibility by Advanced Micro- and Nanospectroscopy Techniques. ACS Sens 2024; 9:4887-4897. [PMID: 39291908 PMCID: PMC11443521 DOI: 10.1021/acssensors.4c01455] [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: 06/16/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Radiotherapy is one of the most common approaches for cancer treatment, especially in the case of peripheral nervous system tumors. As it requires exposure to high doses of ionizing radiation, it is important to look for substances that support efficient reduction of the tumor volume with simultaneous prevention of the surrounding noncancerous cells. Cannabidiol (CBD), which exhibits both anticancer and neuroprotective properties, was applied as a potential modulator of radiological response; however, its influence on cells undergoing irradiation remains elusive. Here, we have applied high-resolution optical spectroscopy techniques to capture biomolecules associated with CBD shielding of normal and damaging cancerous cells upon X-ray exposure. Conventional Raman (RS) and Fourier transformed infrared (FT-IR) spectroscopies provided semiquantitative information mainly about changes in the concentration of total lipids, DNA, cholesteryl esters, and phospholipids in cells. A through assessment of the single cells by atomic force microscopy coupled with infrared spectroscopy (AFM-IR) allowed us to determine not only the alterations in DNA content but also in its conformation due to cell treatment. Pronounced nanoscale changes in cholesteryl ester metabolites, associated with CBD treatment and radiation, were also observed. AFM-IR chemoselective maps of the single cells indicate the modified distribution of cholesteryl esters with 40 nm spatial resolution. Based on the obtained results, we propose a label-free and fast analytical method engaging optical spectroscopy to assess the mechanism of normal and cancerous cell susceptibility to ionizing radiation when pretreated with CBD.
Collapse
Affiliation(s)
- Karolina Chrabąszcz
- Institute of Nuclear Physics Polish
Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| | - Katarzyna Pogoda
- Institute of Nuclear Physics Polish
Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| | - Klaudia Cieżak
- Institute of Nuclear Physics Polish
Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish
Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| | - Wojciech M. Kwiatek
- Institute of Nuclear Physics Polish
Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| |
Collapse
|
245
|
Cheung H, Kang H, Lee HJ, Chung Y, Shin H, Lee S, Kim JH. Homologous Targeting Effect of Cancer Cell-Derived Liposomes (Memposomes) Mediated by Cell Adhesion Molecules: Role of E-cadherin. Biomolecules 2024; 14:1212. [PMID: 39456144 PMCID: PMC11506462 DOI: 10.3390/biom14101212] [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: 07/26/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Cell membrane-derived liposomes, termed Memposomes, serve as promising carriers for drug delivery due to their ability to closely mimic cells and efficiently target specific cells. Liposomes derived from cancer cell membranes, in particular, exhibit homologous targeting capabilities, making them potential candidates for cancer-specific drug delivery. However, the underlying mechanisms and specific proteins responsible for this homologous targeting phenomenon remain debated. This study focuses on the role of E-cadherin, a cell adhesion molecule implicated in homophilic adhesion, in influencing the homologous targeting ability of Memposomes derived from cancer cell membranes. E-cadherin expression patterns were assessed in various cell lines, categorizing them into E-cadherin-positive and -negative groups. Memposomes were produced for each group, and their targeting tendencies were evaluated. This study confirmed that E-cadherin expression significantly influenced the homologous targeting ability of the Memposomes. The cell lines with higher E-cadherin expression levels exhibited a more pronounced homologous targeting effect. This research demonstrates that cell adhesion molecules, particularly E-cadherin involved in homophilic adhesion, play a pivotal role in influencing the cell targeting ability of Memposomes. This study further validates the stability, safety, and purity of Memposomes, emphasizing their potential as effective drug delivery vehicles for the development of cell-specific therapies.
Collapse
Affiliation(s)
- Hyein Cheung
- College of Pharmacy and Bionanocomosite Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Haewon Kang
- College of Pharmacy and Bionanocomosite Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hyo Jung Lee
- Department of Regulatory Science, Institute of Regulatory Innovation through Science, Graduated School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea (H.S.)
| | - Yunjae Chung
- Department of Regulatory Science, Institute of Regulatory Innovation through Science, Graduated School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea (H.S.)
| | - Hanbo Shin
- Department of Regulatory Science, Institute of Regulatory Innovation through Science, Graduated School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea (H.S.)
| | - Sangmin Lee
- College of Pharmacy and Bionanocomosite Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Regulatory Science, Institute of Regulatory Innovation through Science, Graduated School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea (H.S.)
| | - Jong-Ho Kim
- College of Pharmacy and Bionanocomosite Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Regulatory Science, Institute of Regulatory Innovation through Science, Graduated School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea (H.S.)
| |
Collapse
|
246
|
Alarcón-Aldana JS, Visser L, Rueda-Forero NJ, Pinzón-Reyes EH, Rondón-Villarreal P, Suárez-Barrera MO. Enhancing the Cytotoxicity and Apoptotic Efficacy of Parasporin-2-Derived Variants (Mpp46Aa1) on Cancer Cell Lines. Toxins (Basel) 2024; 16:415. [PMID: 39453191 PMCID: PMC11511244 DOI: 10.3390/toxins16100415] [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: 07/30/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Parasporin PS2Aa1, recently renamed Mpp46Aa1, is an anti-cancer protein known for its selectivity against various human cancer cell lines. We genetically modified native PS2Aa1 to create a library of approximately 100 mutants. From this library, we selected promising mutants based on their half-maximal inhibitory concentration (IC50) and sequence variations. In this study, Variant 3-35, with the G257V substitution, demonstrated increased cytotoxicity and selectivity against the colon cancer cell line SW480. Conversely, Variant N65, featuring substitutions N92D, K175R, and S218G, yielded the most favorable results against the cancer cell lines SW-620, MOLT-4, and Jurkat. The caspase 3/7 and 9, Annexin V-Cy3 and 6-GFDA activities, and, most notably, mitochondrial membrane permeabilization assays confirmed the apoptotic marker elevation. These findings indicate that residues 92, 175, 218, and 257 may play a critical role in the cytotoxic activity and selectivity. We successfully obtained genetically improved variants with substitutions at these key amino acid positions. Additionally, we conducted molecular dynamic simulations to explore the potential interactions between PS2Aa1 and the CD59 GPI-anchored protein. The simulation results revealed that residues 57, 92, and 101 were consistently present, suggesting their possible significance in the interactions between parasporin and the CD59 protein.
Collapse
Affiliation(s)
- Juan S. Alarcón-Aldana
- Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación MASIRA, Universidad de Santander, Bucaramanga 680002, Colombia; (J.S.A.-A.); (N.J.R.-F.); (E.H.P.-R.); (P.R.-V.)
| | - Lydia Visser
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9700 AB Groningen, The Netherlands;
| | - Nohora J. Rueda-Forero
- Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación MASIRA, Universidad de Santander, Bucaramanga 680002, Colombia; (J.S.A.-A.); (N.J.R.-F.); (E.H.P.-R.); (P.R.-V.)
| | - Efraín H. Pinzón-Reyes
- Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación MASIRA, Universidad de Santander, Bucaramanga 680002, Colombia; (J.S.A.-A.); (N.J.R.-F.); (E.H.P.-R.); (P.R.-V.)
| | - Paola Rondón-Villarreal
- Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación MASIRA, Universidad de Santander, Bucaramanga 680002, Colombia; (J.S.A.-A.); (N.J.R.-F.); (E.H.P.-R.); (P.R.-V.)
| | - Miguel O. Suárez-Barrera
- Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación MASIRA, Universidad de Santander, Bucaramanga 680002, Colombia; (J.S.A.-A.); (N.J.R.-F.); (E.H.P.-R.); (P.R.-V.)
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9700 AB Groningen, The Netherlands;
| |
Collapse
|
247
|
Wang Y, Wang Y, Jin J. A Graph-Informed Modeling Framework Empowering Gene Pathway Discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614661. [PMID: 39386572 PMCID: PMC11463593 DOI: 10.1101/2024.09.24.614661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
This study introduces a novel graph-informed modeling framework for improving the statistical analysis of gene expression data, particularly in the context of identifying differentially expressed gene pathways and gene expression-assisted disease classification in a high-dimensional data setting. By integrating gene regulatory network information into hypothesis testing for the difference between mean vectors and linear discriminant analysis, we aim to effectively capture and utilize previously validated external gene interaction information. Our method leverages a block-coordinate descent approach which enables us to incorporate mixed graph information into linear structural equation modeling, accommodating directed/undirected edges and potential cycles in gene regulatory networks. Extensive simulations under various data scenarios have demonstrated the effectiveness of our approach with improved power for gene pathway tests and disease classification over existing methods. An application to a lung cancer dataset from the Cancer Genome Atlas Program (TCGA) further exemplifies the potential of our graph-informed approach in empowering the detection of differentially expressed gene pathways and gene expression-based classification of different lung cancer stages. Our findings underscore the potential utility of incorporating gene regulatory network information in gene pathway analysis, setting the stage for future advancements in gene pathway discovery, disease diagnosis, and treatment strategies.
Collapse
|
248
|
Mishra P, Faruqui T, Khanam S, Khubaib M, Ahmad I, Saeed M, Khan S. Sustainable synthesis of bakuchiol-mediated gold nanoparticles for drug delivery against bacterial strains and tumor microenvironments, and its in silico target proteins identification. Front Mol Biosci 2024; 11:1469107. [PMID: 39385982 PMCID: PMC11462060 DOI: 10.3389/fmolb.2024.1469107] [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] [Received: 07/23/2024] [Accepted: 08/16/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction The sustained synthesis of gold nanoparticles (GNPs) has gained significant attention in biomedical applications. In this study, we explored the antibacterial and anticancer potential of bakuchiol-mediated gold nanoparticles (Bak-GNPs). Bakuchiol, a natural compound found in Psoralea corylifolia seeds, serves as both a reducing and stabilizing agent for green synthesis of GNPs. Our objectives include network analysis, molecular docking, synthesis of GNPs, characterization, and antipathogenic and anticancer efficacy of Bak-GNPs against lung and liver cancers. Methods Protein-protein interaction networks were analyzed to identify effective protein targets for bakuchiol in lung and liver cancers. A molecular docking study was performed to validate the efficacy of the target protein against lung and liver cancer. Furthermore, Bak-GNPs were synthesized using bakuchiol and characterized by various techniques such as UV-visible spectroscopy, dynamic light scattering (DLS), zeta potential transmission electron microscopy (TEM), and Fourier-transform infrared (FTIR) spectroscopy, and their potential against pathogens and lung and liver cancers. Results GNAI3 emerged as the most promising target, with a binding energy of -7.5 kcal/mol compared to PTGER3's -6.9 kcal/mol, different characterization techniques revealed the successful synthesis of Bak-GNPs. Bak-GNPs exhibited potent antibacterial activity against both Gram-positive and Gram-negative bacteria, as confirmed by minimum inhibitory concentration (MIC) values. Bak-GNPs demonstrated significant anticancer effects on A549 (lung cancer) and HepG2 (liver cancer) cells, with IC50 values of 11.19 μg/mL and 6.6 μg/mL, respectively. Induction of apoptosis and inhibition of cell proliferation were observed in both the cell lines. The increased production of reactive oxygen species (ROS) contributes to its anticancer effects. Discussion This study highlights promising biomedical applications of bakuchiol-mediated GNPs. This green synthesis approach using bakuchiol provides a sustainable method for producing nanoparticles with enhanced biological activities. Further exploration of the pharmacological properties and mechanisms of Bak-GNPs is required to optimize their therapeutic efficacy for clinical use.
Collapse
Affiliation(s)
- Pooja Mishra
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Tabrez Faruqui
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Sheeba Khanam
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Mohd Khubaib
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Salman Khan
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
249
|
Guevara-Ramírez P, Cadena-Ullauri S, Paz-Cruz E, Ruiz-Pozo VA, Tamayo-Trujillo R, Cabrera-Andrade A, Zambrano AK. Gut Microbiota Disruption in Hematologic Cancer Therapy: Molecular Insights and Implications for Treatment Efficacy. Int J Mol Sci 2024; 25:10255. [PMID: 39408584 PMCID: PMC11476909 DOI: 10.3390/ijms251910255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Hematologic malignancies (HMs), including leukemia, lymphoma, and multiple myeloma, involve the uncontrolled proliferation of abnormal blood cells, posing significant clinical challenges due to their heterogeneity and varied treatment responses. Despite recent advancements in therapies that have improved survival rates, particularly in chronic lymphocytic leukemia and acute lymphoblastic leukemia, treatments like chemotherapy and stem cell transplantation often disrupt gut microbiota, which can negatively impact treatment outcomes and increase infection risks. This review explores the complex, bidirectional interactions between gut microbiota and cancer treatments in patients with HMs. Gut microbiota can influence drug metabolism through mechanisms such as the production of enzymes like bacterial β-glucuronidases, which can alter drug efficacy and toxicity. Moreover, microbial metabolites like short-chain fatty acids can modulate the host immune response, enhancing treatment effectiveness. However, therapy often reduces the diversity of beneficial bacteria, such as Bifidobacterium and Faecalibacterium, while increasing pathogenic bacteria like Enterococcus and Escherichia coli. These findings highlight the critical need to preserve microbiota diversity during treatment. Future research should focus on personalized microbiome-based therapies, including probiotics, prebiotics, and fecal microbiota transplantation, to improve outcomes and quality of life for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Alejandro Cabrera-Andrade
- Escuela de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170124, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170124, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| |
Collapse
|
250
|
Zhang W, Park HB, An EK, Kim SJ, Ryu D, Kim D, Lim D, Hwang J, Kwak M, You S, Lee PCW, Jin JO. Fucoidan from Durvillaea Antarctica enhances the anti-cancer effect of anti-PD-L1 antibody by activating dendritic cells and T cells. Int J Biol Macromol 2024; 280:135922. [PMID: 39322135 DOI: 10.1016/j.ijbiomac.2024.135922] [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: 07/09/2024] [Revised: 09/08/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Immune checkpoint inhibitors are showing groundbreaking results in tumor immunotherapy. However, there are cases where treatment efficiency is insufficient due to limitations in immune activity, and various trials to overcome this are being studied. In this study, we investigated the immune activation ability of fucoidan extracted from Durvillaea antarctica (FDA) and whether it can enhance the anti-cancer effects of immune checkpoint inhibitors. FDA treatment resulted in an elevation of co-stimulator and major histocompatibility complex molecule expression, as well as the production of pro-inflammatory cytokines in bone marrow-derived and splenic dendritic cells (DCs). Administration of 50 mg/kg FDA increased the number of splenic CD8 T cells by >1.4-fold compared to PBS administration. Additionally, 50 mg/kg FDA increased the production of IFN-γ in CD4 and CD8 T cells by 4.3-fold and 7.2-fold, respectively, compared to the PBS control. FDA promoted immune cell activation was TLR4 dependent. Furthermore, anti-PD-L1 antibody administration inhibited CT-26 tumor growth by approximately 3-fold compared to the PBS control group, whereas combined treatment with FDA and anti-PD-L1 antibody showed an 8.4-fold tumor growth inhibition effect compared to the PBS control group. Therefore, FDA may be used to enhance the anti-cancer effects of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Hae-Bin Park
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 05505, South Korea
| | - Dayoung Kim
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Daeun Lim
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Juyoung Hwang
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - SangGuan You
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, 120 Gangneung Daehangno, Gangneung, Gangwon 210-702, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, 05505, South Korea
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| |
Collapse
|