1
|
Julappagari M, Haque S, Tripathy S, Londhe S, Patel A, Banerjee R, Patra CR. Gold nanoparticles-based targeted delivery of rapamycin and Olaparib to breast cancer: An in vitro and in vivo approach. Bioorg Chem 2025; 158:108322. [PMID: 40073595 DOI: 10.1016/j.bioorg.2025.108322] [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: 11/30/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
Triple negative form of breast cancer (abbreviated as TNBC) is considered as the most aggressive form causing high mortality worldwide. Different treatment modalities such as chemotherapy, surgery, hormonal therapy and radiation therapy are employed for eliminating breast cancer, which are associated with many limitations. Therefore, considering the significance of metal nanoparticles in the biomedical sector, especially gold nanoparticles, in the current manuscript, we have designed and developed a combinatorial approach for synthesizing two types of gold (Au) nanoformulations (Au-Dex-MUA-Rapa, Au-Dex-MUA-Ola) using 11-mercaptoundecanoic acid (MUA), dexamethasone (Dex) (glucocorticoid receptor targeted molecule) along with rapamycin (Rapa: inhibitor of mTOR) or olaparib (Ola: inhibitor of PARP) against TNBC. These gold nanoformulations were characterized thoroughly using several analytical techniques such as TEM, spectroscopy, DLS, HPLC and ICPOES. The in vitro MTT assays (normal cells: HEK-293 and CHO) and ex vivo CAM assay displays the biocompatible properties of the conjugated gold nanoformulations. Further, the anticancer properties of the conjugated gold nanoformulations in TNBC cells (MDA-MB-231) were evaluated through several in vitro experiments along with plausible mechanism of action. The intraperitoneal administration of gold nanoformulations into the breast tumor bearing BALB/c mice inhibits the tumor growth and increases their survivability. Additionally, we have investigated the plausible mechanistic studies behind the anticancer properties of the conjugated gold nanoformulations. Finally, we have found the non-toxic nature of these nanoformulations at therapeutic dose. Considering the above results, the conjugated gold nanoformulations could be used as an alternative therapeutic strategy for the treatment of breast carcinoma in near future.
Collapse
Affiliation(s)
- Mamatha Julappagari
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
| | - Shagufta Haque
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sanchita Tripathy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Swapnali Londhe
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arti Patel
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
| | - Rajkumar Banerjee
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Pontoriero A, Critelli P, Zeppieri M, Bosurgi A, Guercio S, Caffo M, Angileri FF, Parisi S, Lavalle S, Pergolizzi S. Nano-drug delivery systems integrated with low radiation doses for enhanced therapeutic efficacy in cancer treatment. World J Clin Cases 2025; 13:101719. [DOI: 10.12998/wjcc.v13.i10.101719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/11/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Precision medicine is an emerging field that includes tumor-targeted delivery and tumor microenvironment. This review explores the synergistic potential of combining nano-drug delivery systems with low radiation doses to achieve optimized therapeutic outcomes, particularly in the context of cancer treatment. Nanoparticle-based drug carriers offer precise and targeted delivery, enhancing the therapeutic index of anticancer agents. The use of lower radiation doses has become a focus in radiation oncology to minimize off-target effects on healthy tissues in palliation treatment with high-target volume lesions.
AIM To conduct a bibliometric review of nanomedicine and glioblastoma (GBM), all relevant studies from the last two decades were included.
METHODS The search strategy comprised the keywords ”nanomedicine “and “glioblastoma” in the title and/or abstract. All English-language documents from 1 January 2000 to 31 December 2023 were considered for the analysis. R code (version 4.2.0) with R Studio (version 2022.12.0-353) and the Bibliometrix package (version 4.0.1) were used for the analysis. A total of 680 documents were collected.
RESULTS We analyzed the bibliometric features of nanomedicine in glioma. With the limitations of the research, our analysis aims to highlight the increasing interest of researchers in the precision medicine field in GBM treatment and lead us to suggest further studies focusing on the association between nanomedicine and radiotherapy.
CONCLUSION Due to the poor prognosis associated with GBM, new therapeutic approaches are necessary. There is an increasing interest in precision medicine, which includes nanomedicine and radiotherapy, for GBM treatment. This integration enhances the efficacy of targeted treatments and provides a promising avenue for reducing adverse effects, signifying a notable advancement in precision oncology.
Collapse
Affiliation(s)
- Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Paola Critelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Alberto Bosurgi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Stefania Guercio
- Neurosurgery Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, Messina, Italy, Messina 98125, Italy
| | - Maria Caffo
- Neurosurgery Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, Messina, Italy, Messina 98125, Italy
| | - Filippo Flavio Angileri
- Neurosurgery Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, Messina, Italy, Messina 98125, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Salvatore Lavalle
- Department of Medicine and Surgery, University of Enna "Kore", Enna 94100, Italy
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| |
Collapse
|
3
|
Poonia N, Kumar V, Subudhi RN, Dalabehera M, Setia A, Bora KS, Arora V. Iron oxide nanoparticles: a versatile nanoplatform for the treatment and diagnosis of ovarian cancer. Ther Deliv 2025; 16:379-392. [PMID: 39722582 DOI: 10.1080/20415990.2024.2442301] [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: 06/03/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
Ovarian cancer remains one of the main causes of human mortality, accounting for millions of deaths every year. Despite of several clinical options such as chemotherapy, photodynamic therapy (PDT), hormonal treatment, radiation therapy, and surgery to manage this disease, the mortality rate is still very high. This alarming statistic highlights the urgent need for innovative approaches to improve both diagnosis and treatment. Success stories of iron oxide nanoparticles, i.e. Ferucarbotran (Resovist®) and Ferrixan (Cliavist®) for liver imaging, CNS (Central nervous system) imaging, cell labeling, etc. have motivated researchers to explore these nanocarriers for treatment and diagnosis of different diseases. Iron oxide nanoparticles have improved the therapeutic efficacy of anticancer drugs through targeted delivery, heat/ROS (reactive oxygen species) generation on application of external energy and have also shown great potential as contrast agents for magnetic resonance imaging (MRI). Their unique magnetic properties enable sensitive imaging, and surface modification allows the attachment of specific biomolecules for targeted detection of ovarian cancer cells. Their unique properties, viz. magnetic responsiveness and surface functionalization, make them versatile tools for enhancing both imaging and therapeutic outcomes. Present article reviews the literature on the synthesis, functionalization, and applications of iron oxide nanoparticles in management of ovarian cancer.
Collapse
Affiliation(s)
- Neelam Poonia
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| | - Vipan Kumar
- Department of Pharmaceutical Chemistry, Swami Vivekanand College of Pharmacy, Rajpura, India
| | | | - Manoj Dalabehera
- University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| | - Anupama Setia
- Department of Pharmaceutics, JCDM College of Pharmacy, Sirsa, India
| | - Kundan Singh Bora
- Department of Pharmacognosy, University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| | - Vimal Arora
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| |
Collapse
|
4
|
Sigler GI, Murtha J, Varley PR. Diagnostic Advances and Novel Therapeutics in Peritoneal Metastasis. Surg Oncol Clin N Am 2025; 34:173-194. [PMID: 40015798 DOI: 10.1016/j.soc.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Appropriate assessment of disease burden in patients with peritoneal surface malignancy (PSM) is critical for treatment decision-making, and conventional cross-sectional imaging (computed tomography and/or MRI) often underestimates burden of disease. Advances in imaging for PSM include novel functional imaging modalities that target cells unique to the tumor microenvironment. Novel alternative methods of diagnosis and disease monitoring are also potentially applicable to management of PSM. These include forms of "liquid biopsy" targeting circulating tumor DNA. Novel regional therapies include both new therapeutic agents (immune-based and nanoparticle-based), as well as new methods of delivery such as pressurized intraperitoneal aerosolized chemotherapy.
Collapse
Affiliation(s)
- Gregory I Sigler
- Division of Surgical Oncology, Department of General Surgery, Complex General Surgical Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Jacqueline Murtha
- Department of General Surgery, General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Patrick R Varley
- Division of Surgical Oncology, Department of General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA; William S. Middleton Memorial Veterans Affairs Hospital, Madison, WI, USA.
| |
Collapse
|
5
|
Chota A, Abrahamse H, George BP. Chemotoxic and phototoxic effects of liposomal co-delivery of green synthesized silver nanoparticles and ZnPcS 4 for enhanced photodynamic therapy in MCF-7 breast cancer cells: An in vitro study. Biomed Pharmacother 2025; 185:117986. [PMID: 40090284 DOI: 10.1016/j.biopha.2025.117986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
Breast cancer remains a significant challenge in oncology, despite notable advances in treatment methods. Traditional therapies such as surgery, chemotherapy, radiation, and hormonal treatments have long been used to manage breast cancer. However, often patients experience treatment failure, resulting in disease recurrence and progression. Therefore, this study explores the therapeutic potential of green-synthesized silver nanoparticles (AgNPs), using the root methanol (MeOH) extract of the African medicinal plant Dicoma anomala (D. anomala) as a reducing agent, to combat breast cancer. AgNPs were synthesized using a bottom-up approach and later modified with liposomes (Lip) loaded with the photosensitizer zinc phthalocyanine tetrasulfonate (Lip@ZnPcS4) through the thin film hydration method. Prior to in vitro cell culture studies, UV-Vis spectroscopy was used to study the in vitro drug release kinetics of nanoparticles (NPs) at pH 5.8 and 7.4 respectively. After a 24 h treatment period, MCF-7 breast cancer cells were evaluated for cell cytotoxicity using lactate dehydrogenase Cyto-Tox96® Non-Radioactive Cytotoxicity Assay Kit LDH and cell viability using the CellTiter-Glo® ATP luminescence assay kit. Cell death studies were analyzed using an inverted light microscope for morphological changes, fluorescence microscopy for reactive oxygen species (ROS) detection and Live/Dead cell viability, human p53 protein analysis using enzyme-linked immunosorbent assay (ELISA), apoptotic and anti-apoptotic protein analysis by immunofluorescence, and gene expression analysis using real-time reverse transcription polymerase chain reaction (RT-PCR) assay. The experiments were conducted in quadruplicate (n = 4), and the results were analyzed using IBM SPSS statistical software version 27, with a 95 % confidence interval. The synthesized NPs and nanocomplexes, including AgNPs, AgNPs-Lip, Lip@ZnPcS4, and AgNPs-Lip@ZnPcS4, demonstrated significant cytotoxicity and therapeutic potential against MCF-7 breast cancer cells. Notably, apoptosis was induced, primarily through the activation of the intrinsic pathway. Given the difficult prognosis associated with breast cancer, these findings highlight the promise of liposomal nanoformulations (NFs) in cancer photodynamic therapy (PDT), supporting further investigation in in vivo settings.
Collapse
Affiliation(s)
- Alexander Chota
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa.
| |
Collapse
|
6
|
El Halawany M, Sharaky M, Aziz D. Stearyl amine tailored spanlastics embedded within tetronic ® nanogel for boosting the repurposed anticancer potential of mebendazole: formulation, in vitro profiling, cytotoxicity assessment, and in vivo permeation analysis. Daru 2025; 33:17. [PMID: 40156679 PMCID: PMC11954785 DOI: 10.1007/s40199-025-00560-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/06/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Mebendazole (MBZ) is an anthelmintic drug that was repurposed as an anti-cancer agent. OBJECTIVES This study aimed at formulating MBZ into stearylamine tailored spanlastics dispersed in nanogel for enhancing MBZ anti-tumor efficacy against skin cancer. METHODS MBZ spanlastics were prepared by thin film hydration using 21 × 31 factorial design. The formulation variables were the total amount (mg) of Span 60 and Tween 80 in the formulations and the ratio between Span 60 and Tween 80. RESULTS Optimal spanlastics formulation was composed of 400 mg of Span 60 and Tween 80 in a ratio of 2:1 and showed EE% of 78 ± 2.9% and PS of 284.00 ± 35.36 nm. Stearylamine (20 mg) was added to the optimized formulation and showed acceptable positive charge (zeta potential = 47.53 ± 1.50 mV). It was dispersed in 30% Tetronic®1107 solution to form a nanogel. MBZ nanogel was assessed for their cytotoxic effect on cell proliferation against human malignant melanoma and epidermoid carcinoma cell lines and showed 38.70 ± 1.70% and 48.60 ± 0.50% (respectively) cell proliferation compared to the control group (100%). Finally, its permeation through Wistar rat skin was tested. CONCLUSION SA-spanlastics nanogel holds potential as an effective nanocarrier for boosting MBZ anti-cancer efficacy.
Collapse
Affiliation(s)
- Mai El Halawany
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street 11562, Cairo, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Kasr El-Ainy Street 11562, Cairo, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6Th October City 12573, Giza, Egypt
| | - Diana Aziz
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street 11562, Cairo, Egypt.
| |
Collapse
|
7
|
Fu Z, Wang S, Zhou X, Ouyang L, Chen Z, Deng G. Harnessing the Power of Traditional Chinese Medicine in Cancer Treatment: The Role of Nanocarriers. Int J Nanomedicine 2025; 20:3147-3174. [PMID: 40103746 PMCID: PMC11913986 DOI: 10.2147/ijn.s502104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
For centuries, traditional Chinese medicine (TCM) has had certain advantages in the treatment of tumors. However, due to their poor water solubility, low bioavailability and potential toxicity, their effective delivery to target sites can be a major challenge. Nanocarriers based on the active ingredients of TCM, such as liposomes, polymer nanoparticles, inorganic nanoparticles, and organic/inorganic nanohybrids, are a promising strategy to improve the delivery of TCM, resulting in higher therapeutic outcomes and fewer side effects. Therefore, this article intends to review the application of Chinese medicine nano preparation in tumor. Firstly, we introduce the classification and synthesis of nanometer preparations of Chinese medicine. The second part mainly introduces the different responses of TCM nano-preparations in the course of treatment to introduce how TCM nano-preparations play a role in anti-tumor therapy. The third part focuses on Different response modes of Chinese medicine nano preparations in tumor therapy. The fourth part elucidates the application of Chinese medicine nano preparations in the treatment of cancer. Finally, the research direction to be explored in related fields is put forward.
Collapse
Affiliation(s)
- Ziyu Fu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People's Republic of China
| | - Shengmei Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People's Republic of China
| | - Xin Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People's Republic of China
| | - Linqi Ouyang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People's Republic of China
| | - Zhen Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People's Republic of China
| | - Guiming Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People's Republic of China
- The second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410005, People's Republic of China
| |
Collapse
|
8
|
Li X, Liu Y, Wu X, Huang R, Chen S, Yuan K. Ultrasound meets nanomedicine: towards disease treatment and medical imaging. Mikrochim Acta 2025; 192:215. [PMID: 40053162 DOI: 10.1007/s00604-025-07042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/11/2025] [Indexed: 03/18/2025]
Abstract
As a kind of mechanical wave, ultrasound has been widely employed in the biomedical field due to its superiors of deep tissue penetration, non-destructiveness and non-toxicity. In this review, we highlight current progress and prospects in using ultrasound as a powerful tool for disease treatment and medical imaging, including (1) ultrasound as energy input for driving nano/micromotor in drug delivery, this part first introduces the synthesis and motion behavior of nano/micromotors, then reviews the small molecular and macromolecular compounds that the nano/micromotors are delivering; (2) sonosensitive nanomaterials for disease therapy, the sonodynamic, sonopiezoelectric, sonothermal, and sonomechanical therapy will all be covered; (3) ultrasound as a non-invasive technique for nano/micromotor tracking or medical imaging; (4) the sonoporation of nano/microbubble. Future challenges in using ultrasound for disease treatment or medical imaging will also be described in the conclusion part.
Collapse
Affiliation(s)
- Xiaochun Li
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yanting Liu
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Xuewan Wu
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Rui Huang
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Shaoqi Chen
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Kaisong Yuan
- Department of Ultrasound, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Bio-Analytical Laboratory, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
9
|
Yuan Y, Beilharz S, Everson HR, Nupnar N, Debnath MK, Vinella D, Urueña JM, Örge FH, Hore MJA, Mathur D, Karayilan M. Injectable Fluorescent Bottlebrush Polymers for Interventional Procedures and Biomedical Imaging. Biomacromolecules 2025; 26:1234-1250. [PMID: 39849929 DOI: 10.1021/acs.biomac.4c01550] [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: 01/25/2025]
Abstract
Injectable biomaterials play a vital role in modern medicine, offering tailored functionalities for diverse therapeutic and diagnostic applications. In ophthalmology, for instance, viscoelastic materials are crucial for procedures such as cataract surgery but often leave residues, increasing postoperative risks. This study introduces injectable fluorescent viscoelastics (FluoVs) synthesized via one-step controlled radical copolymerization of oligo(ethylene glycol) acrylate and fluorescein acrylate. These bottlebrush-shaped polymers exhibit enhanced fluorescence intensity for improved traceability and facile removal postsurgery. To prevent aggregation, charged terpolymers were synthesized, ensuring intra- and intermolecular electrostatic repulsion. Dynamic light scattering and energy-conserved dissipative particle dynamics simulations revealed how the fluorescein content and monomer sequence affect the hydrodynamic size of these copolymers. Biocompatibility assessments showed that FluoVs maintained cell viability comparable to commercial hydroxypropyl methylcellulose and nonfluorescent poly(oligo(ethylene glycol) acrylate) controls. The FluoVs combine high fluorescence intensity, low viscosity, and excellent biocompatibility, offering intraoperative traceability and significant advancements for ocular and bioimaging applications.
Collapse
Affiliation(s)
- Yichun Yuan
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Sophia Beilharz
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Heather R Everson
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Nehal Nupnar
- Department of Macromolecular Science and Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Mithun Kumar Debnath
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Daniele Vinella
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Juan Manuel Urueña
- NSF BioPACIFIC MIP, University of California Santa Barbara, Elings Hall, Mesa Road, Santa Barbara, California 93106, United States
| | - Faruk H Örge
- Center for Pediatric Ophthalmology and Adult Strabismus Rainbow Babies and Children's Hospital, University Hospitals Eye Institute, and Department of Ophthalmology and Pediatrics, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Michael J A Hore
- Department of Macromolecular Science and Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Divita Mathur
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Metin Karayilan
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
10
|
Alqurashy NN, Yousef MI, Hussein AA, Kamel MA, El Wakil A. Monascus red pigment influence on hydroxyapatite nanoparticles-mediated renal toxicity in rats. Sci Rep 2025; 15:2715. [PMID: 39837868 PMCID: PMC11750980 DOI: 10.1038/s41598-024-84959-z] [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: 11/22/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
Hydroxyapatite nanoparticles (HANPs) have been applied in several biomedical fields. However, its interaction with biological systems is less exploited. This study aimed to characterize HANPs, examine their influence on kidneys, and explore the potential protective effects of naturally extracted red pigment (RP) from Monascus purpureus against HANPs-induced renal toxicity. To this aim, forty eight adult male rats were randomly divided into 8 equal groups: a control group receiving 4% dimethyl sulfoxide (the solvent for HANPs), three groups receiving extracted RP at different doses of 10, 20, and 40 mg/kg, a group receiving HANPs at a dose of 88.3 mg/kg, and three more groups receiving a double treatment of HANPs associated with RP. The respective treatment was given daily by oral gavage to animals for 50 days which is the duration of the whole experiment. The renal toxicity caused by HANPs was manifested by aberrations in kidney function parameters, intensification of oxidative stress markers, and a decrease in the activity of antioxidant enzymes. Moreover, an increase in inflammatory (TNF-α and TGF-β) and apoptotic (caspace-3) markers, an elevation in gene-based kidney injuries markers (Kim-1 and lipocalin-2), and pathological tissue changes were determined. Meanwhile, co-treatment with different doses of biopigment and HANPs have reduced oxidative stress via their potent antioxidant effect. This was confirmed by pronounced improvement in the measured parameters along with the histological structural enhancement in a dose dependent manner compared to controls. To sum up, RP from M. purpureus has potential protective benefits in mitigating the adverse effects of HANPs.
Collapse
Affiliation(s)
- Nasser N Alqurashy
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Mokhtar I Yousef
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Ahmed A Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Abeer El Wakil
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Alexandria, 21526, Egypt.
| |
Collapse
|
11
|
Kumar N, Mangla M. Nanotechnology and nanobots unleashed: pioneering a new era in gynecological cancer management - a comprehensive review. Cancer Chemother Pharmacol 2025; 95:18. [PMID: 39754614 DOI: 10.1007/s00280-024-04747-4] [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: 08/28/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
INTRODUCTION Gynecological cancers, such as ovarian, cervical, and endometrial malignancies, are notoriously challenging due to their intricate biology and the critical need for precise diagnostic and therapeutic approaches. In recent years, groundbreaking advances in nanotechnology and nanobots have emerged as game-changers in this arena, offering the promise of a new paradigm in cancer management. This comprehensive review delves into the revolutionary potential of these technologies, showcasing their ability to transform the landscape of gynecological oncology. METHODOLOGY A systematic literature search spanning from March 2005 to August 2024 was conducted using major databases such as PubMed, Google Scholar, and Scopus. Keywords included "nanotechnology," "nanobots," "gynecological cancers," "ovarian cancer," "cervical cancer," and "endometrial cancer." Relevant articles published in English were selected based on their focus on nanotechnology and nanobots in the diagnosis, treatment, and management of gynecological cancers. The findings were synthesized to present a coherent overview of how nanotechnology and nanobots are reshaping gynecological cancer management. The review highlights key innovations, current applications, and future directions for research and clinical implementation. CONCLUSION The integration of nanotechnology and nanobots in gynecological cancer management represents a groundbreaking shift in the field. Recent advancements in nanoscale materials and robotic technology offer unprecedented opportunities for precision diagnosis, targeted drug delivery, and innovative therapeutic approaches. Despite promising developments, challenges such as biocompatibility, safety, and regulatory issues remain. Continued research and clinical trials are essential to overcome these hurdles and fully realize the potential of nanotechnology and nanobots.
Collapse
Affiliation(s)
- Naina Kumar
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India.
| | - Mishu Mangla
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| |
Collapse
|
12
|
Nasrine A, Mohanto S, Narayana S, Ahmed MG. Enhanced pharmacokinetic approach for anastrozole in macromolecule-based silk fibroin nanoparticles incorporated in situ injectables for estrogen-positive breast cancer therapy. J Drug Target 2025:1-28. [PMID: 39754343 DOI: 10.1080/1061186x.2024.2449486] [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: 10/05/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
Breast cancer (BC) is a substantial reason for cancer-related mortality among women across the globe. Anastrozole (ANS) is an effective orally administered hormonal therapy for estrogen+ (ER+) BC treatment. However, several side effects and pharmacokinetic limitations restricted its uses in BC treatment. Therefore, this study developed an in situ gelling injectable-loaded silk fibroin (SF)-ANS NPs, which offers sustained drug release and improved pharmacokinetic properties compared to conventional oral formulations. The optimized in situ gel (ISG) incorporated SF-ANS-NPs were developed, and the pharmacokinetic parameters were accessed in subcutaneous administration of NMU-induced Wistar albino rats. The results demonstrated that SF-ANS-NP-ISG exhibited a significantly higher Cmax, Tmax, and AUC compared to pure ANS suspension. In addition, tumor multiplicity (1.40 ± 0.66), tumor latency (75 ± 9.2 days), and incidence rate (90 ± 2.1%) were recorded, and post-treatment analysis reported a marked reduction in tumor volume and weight compared to positive control within 90 days of a single dose. The SF-ANS-NP-ISG treated group's histopathological assessment indicated a low-grade carcinoma, reduced epithelial hyperplasia, and haemorrhage in mammary tumor tissues compared to positive control. Thus, the SF-ANS-NPs-ISG investigated to overcome the pharmacokinetic limitations of ANS further exhibited targeted delivery and bioavailability compared to conventional techniques.
Collapse
Affiliation(s)
- Arfa Nasrine
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Harohalli, Kanakpura Road, Bangalore, Karnataka, 562112, India
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, 575018, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, 575018, India
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, 575018, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, 575018, India
| |
Collapse
|
13
|
Deng B, Ke B, Tian Q, Gao Y, Zhai Q, Zhang W. Targeting AURKA with multifunctional nanoparticles in CRPC therapy. J Nanobiotechnology 2024; 22:803. [PMID: 39734237 DOI: 10.1186/s12951-024-03070-7] [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: 08/04/2024] [Accepted: 12/11/2024] [Indexed: 12/31/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) presents significant therapeutic challenges due to its aggressive nature and poor prognosis. Targeting Aurora-A kinase (AURKA) has shown promise in cancer treatment. This study investigates the efficacy of ART-T cell membrane-encapsulated AMS@AD (CM-AMS@AD) nanoparticles (NPs) in a photothermal-chemotherapy-immunotherapy combination for CRPC. Bioinformatics analysis of the Cancer Genome Atlas-prostate adenocarcinoma (TCGA-PRAD) dataset revealed overexpression of AURKA in PCa, correlating with poor clinical outcomes. Single-cell RNA sequencing data from the GEO database showed a significant reduction in immune cells in CRPC. Experimentally, T cell membrane-biomimetic NPs loaded with the AURKA inhibitor Alisertib and chemotherapy drug DTX were synthesized and characterized by dynamic light scattering and transmission electron microscopy, showing good stability and uniformity (average diameter: 158 nm). In vitro studies demonstrated that these NPs inhibited CRPC cell proliferation, increased the G2/M cell population, and elevated apoptosis, confirmed by γH2AX expression. In vivo, CM-AMS@AD NPs accumulated in tumor tissues, significantly slowed tumor growth, decreased proliferation, increased apoptosis, and improved the immune environment, enhancing dendritic cell (DC) maturation and increasing CD8 + /CD4 + ratios. These findings suggest that CM-AMS@AD NPs offer a promising triple-combination therapy for CRPC, integrating photothermal, chemotherapy, and immunotherapy, with significant potential for future clinical applications.
Collapse
Affiliation(s)
- Bin Deng
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
- Department of Science and Technology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Binghu Ke
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Qixing Tian
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Yukui Gao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China.
| | - Qiliang Zhai
- Department of Urology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, China.
| | - Wenqiang Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China.
| |
Collapse
|
14
|
Ma L, Kim MO. Advances in Preventive and Therapeutic Strategies for Oral Cancer: A Short Review. J Cancer Prev 2024; 29:113-119. [PMID: 39790224 PMCID: PMC11706729 DOI: 10.15430/jcp.24.027] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Oral cancer is a major global health concern, with high incidence and mortality rates, especially in high-risk populations. Early diagnosis remains a challenge, and current treatments, such as surgery, radiation, and chemotherapy, have limited effectiveness, particularly in advanced stages. Recent advances in targeted therapies and immunotherapy offer promising alternatives, providing more precise and personalized treatment options. Targeted therapies, such as epidermal growth factor receptor inhibitors, aim to disrupt specific molecular pathways in tumor growth, while immunotherapies, including immune checkpoint inhibitors and chimeric antigen receptor-T cell therapy, enhance the body's immune response to fight cancer. Combination therapies, integrating both targeted and immune strategies, are being explored to overcome the limitations of single-agent treatments. This review highlights the current strategies in the prevention and treatment of oral cancer, discusses emerging therapies, explores future research directions, focusing on optimizing existing treatments, identifying new biomarkers, and developing innovative therapeutic approaches. The potential of personalized medicine and combination therapies offers new hope for improving survival rates and quality of life for oral cancer patients.
Collapse
Affiliation(s)
- Lei Ma
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| |
Collapse
|
15
|
Cao X, Feng N, Huang Q, Liu Y. Nanoscale Metal-Organic Frameworks and Nanoscale Coordination Polymers: From Synthesis to Cancer Therapy and Biomedical Imaging. ACS APPLIED BIO MATERIALS 2024; 7:7965-7986. [PMID: 38382060 DOI: 10.1021/acsabm.3c01300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Recently, there has been significant interest in nanoscale metal-organic frameworks (NMOFs) characterized by ordered crystal structures and nanoscale coordination polymers (NCPs) featuring amorphous structures. These structures arise from the coordination interactions between inorganic metal ions or clusters and organic ligands. Their advantages, such as the ability to tailor composition and structure, efficiently encapsulate diverse therapeutic or imaging agents within porous frameworks, inherent biodegradability, and surface functionalization capability, position them as promising carriers in the biomedical fields. This review provides an overview of the synthesis and surface modification strategies employed for NMOFs and NCPs, along with their applications in cancer treatment and biological imaging. Finally, future directions and challenges associated with the utilization of NMOFs and NCPs in cancer treatment and diagnosis are also discussed.
Collapse
Affiliation(s)
- Xianghui Cao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Nana Feng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Qingqing Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| |
Collapse
|
16
|
Ali A, Alamri A, Hajar A. NK/DC crosstalk-modulating antitumor activity via Sema3E/PlexinD1 axis for enhanced cancer immunotherapy. Immunol Res 2024; 72:1217-1228. [DOI: https:/doi.org/10.1007/s12026-024-09536-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/29/2024] [Indexed: 01/06/2025]
|
17
|
Ali A, Alamri A, Hajar A. NK/DC crosstalk-modulating antitumor activity via Sema3E/PlexinD1 axis for enhanced cancer immunotherapy. Immunol Res 2024; 72:1217-1228. [PMID: 39235526 DOI: 10.1007/s12026-024-09536-y] [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: 06/14/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
The complex relationship between natural killer (NK) cells and dendritic cells (DCs) within the tumor microenvironment significantly impacts the success of cancer immunotherapy. Recent advancements in cancer treatment have sought to bolster innate and adaptive immune responses through diverse modalities, aiming to tilt the immune equilibrium toward tumor elimination. Optimal antitumor immunity entails a multifaceted interplay involving NK cells, T cells and DCs, orchestrating immune effector functions. Although DC-based vaccines and NK cells' cytotoxic capabilities hold substantial therapeutic potential, their interaction is frequently hindered by immunosuppressive elements such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells. Chemokines and cytokines, such as CXCL12, CCL2, interferons, and interleukins, play crucial roles in modulating NK/DC interactions and enhancing immune responses. This review elucidates the mechanisms underlying NK/DC interaction, emphasizing their pivotal roles in augmenting antitumor immune responses and the impediments posed by tumor-induced immunosuppression. Furthermore, it explores the therapeutic prospects of restoring NK/DC crosstalk, highlighting the significance of molecules like Sema3E/PlexinD1 in this context, offering potential avenues for enhancing the effectiveness of current immunotherapeutic strategies and advancing cancer treatment paradigms. Harnessing the dynamic interplay between NK and DC cells, including the modulation of Sema3E/PlexinD1 signaling, holds promise for developing more potent therapies that harness the immune system's full potential in combating cancer.
Collapse
Affiliation(s)
- Awais Ali
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, 23200, Pakistan.
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Azraida Hajar
- Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
18
|
Lu W, Li Y, Zhang X, Wang N, Chen D, Zhao Y, Li G, Shi X, Ma X, Su X, Wang F, Shu C, Chen K. Dual-modal overcoming of physical barriers for improved photodynamic cancer therapy via soft organosilica nanocapsules. J Nanobiotechnology 2024; 22:734. [PMID: 39593093 PMCID: PMC11600580 DOI: 10.1186/s12951-024-02945-z] [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] [Accepted: 10/20/2024] [Indexed: 11/28/2024] Open
Abstract
Amidst the burgeoning field of cancer nanomedicine, dense extracellular matrices and anomalous vascular structures in the tumor microenvironment (TME) present substantial physical barriers to effective therapeutic delivery. These physical barriers hinder the optimal bioavailability of nanomedicine. Here, we propose a pioneering dual-modal strategy for overcoming physical barriers via soft organosilica nanocapsules (SMONs). Hyaluronidase-modified flexible spheres work by degrading the extracellular matrix and utilizing their flexible characteristics to enhance penetration into deeper layers. Compared with their stiff counterparts, the SMONs show diminished Young's modulus, then the inherent softness of the SMONs confers distinct advantages, and significantly augmented cellular internalization within 4T1 cells, leading to an amplified in vitro photodynamic therapeutic effect. Furthermore, hyaluronidase-functionalized SMONs (SMONs-HAase) exhibit enhanced tumor penetration in 3D spheroids. Post incorporation of the photosensitizer chlorin e6, when administered intravenously, these soft organosilica nanocapsules amplify the efficacy of photodynamic therapy. In addition, RNA-seq analysis of SMONs-HAase-Ce6 shows it alters gene expression, degrading the extracellular matrix and impairing mitochondrial function. To sum up, this work elucidates the potential of a dual-modal strategy, highlighting the promise of SMONs in overcoming TME physical barriers and optimizing therapeutic outcomes.
Collapse
Affiliation(s)
- Wei Lu
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, P.R. China
| | - Xiaojun Zhang
- Department of Radiology, Children's Hospital of Nanjing Medical University, Nanjing, 210018, P.R. China
| | - Ning Wang
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Dong Chen
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Yatong Zhao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 213161, P.R. China
| | - Guang Li
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410000, P.R. China
| | - Xuzhi Shi
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Xiaobo Ma
- College of Electronic and Optical Engineering & College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Feng Wang
- Department of Radiology, Children's Hospital of Nanjing Medical University, Nanjing, 210018, P.R. China.
| | - Chuqiang Shu
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410000, P.R. China.
| | - Kun Chen
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P.R. China.
| |
Collapse
|
19
|
Li B, Zhao Q, Yang H, Wang X, Zhang Z, Gong Y, Wan X. Long-Circulating and Targeted Liposomes Co-loading Cisplatin and Mifamurtide: Formulation and Delivery in Osteosarcoma Cells. AAPS PharmSciTech 2024; 25:272. [PMID: 39592553 DOI: 10.1208/s12249-024-02992-7] [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: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Osteosarcoma (OS) is one of the most common primary bone sarcoma with high malignant degree and poor prognosis, for which there is an urgent need to develop novel therapeutic approaches. Recent research has revealed that mifamurtide significantly improved the outcome of OS patients when combined with adjuvant chemotherapy drugs including cisplatin (DDP). The present study aimed to construct a drug delivery system co-loading DDP and mifamurtide. Long-circulating targeted liposomes co-loading DDP and mifamurtide were constructed with Soy lecithin (SPC), cholesterol (Chol) and 1,2-distearoylglycero-3-phosphoethanolamine-n-[poly(ethyleneglycol)] (DSPE-PEG), modified with MMP14 targeting peptide BCY-B in the surface of liposomes. In addition to characterization, the cellular uptake, endocytosis pathway and inhibition on cell viability, migration, invasion and cell apoptosis of MG-63 cells were explored. The constructed liposomal delivery possessed the basic characteristics of liposomes and showed high affinity to MG-63 cells, resulting in high uptake efficiency in MG-63 cells. The endocytosis might be involved in multiple pathways including caveolae-mediated endocytosis, clathrin-mediated endocytosis and macropinocytosis, dependently on energy. The constructed long-circulating targeted liposomes co-loading DDP and mifamurtide significantly inhibited the cell viability, migration, invasion and cell apoptosis of MG-63 cells, improving the antitumor effect of DDP and mifamurtide in vitro. The constructed liposomal delivery system is suitable for co-loading DDP and mifamurtide to achieve active tumor targeting, supplying a new strategy for the treatment of OS.
Collapse
Affiliation(s)
- Bo Li
- Department of Musculoskeletal Tumor, Gansu Provincial Hospital, Lanzhou, China
| | - Qianhui Zhao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Hanyu Yang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Xueyuying Wang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Zhijun Zhang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China.
| | - Xu Wan
- Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
20
|
Zhao YC, Li X, Wang CQ, Jiao Y, Shen YN, Wang TJ, Zhang CH. Unveiling the Hidden Risks: An Update Decade-Long Analysis of Abraxane-Related Adverse Events from the FAERS Database. Int J Nanomedicine 2024; 19:11847-11858. [PMID: 39558918 PMCID: PMC11572439 DOI: 10.2147/ijn.s490400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose Abraxane (nanoparticle albumin-bound paclitaxel) is a chemotherapeutic employed commonly for the management of various cancers including breast cancer, non-small cell lung cancer, and pancreatic adenocarcinoma. Although it has clinically beneficial properties, Abraxane is accompanied by multiple adverse events (AEs) that require close observation. This study aims to evaluate the AE profile of Abraxane using recently available data from January 2004 through December 2023 in the FDA Adverse Event Reporting System (FAERS). Patients and Methods The data for Abraxane-related AEs were obtained from the FAERS database. The dataset consisted of patient demographic characteristics as well as information on the types and outcomes of AEs reported. Reporting odds ratios (ROR) as well as proportional reporting ratio (PRR), considering the used definition of anti-cancer agent and AEs, were calculated to investigate any association with Abraxane. Results A total of 10,310 reports associated with Abraxane AEs were identified. Blood and lymphatic system disorders were the most frequent (ROR 6.44), followed by hepatobiliary (ROR 3.16), infections (ROR 1.45), and gastrointestinal disorders (ROR 1.42). Serious outcomes included hospitalization in 36.35% and death in 29.76% of cases. The top adverse reactions matched known profiles, including peripheral sensory neuropathy (ROR: 49.48). The analysis also found new adverse reactions, such as scleroderma-like reactions (ROR: 95.4) and vascular pseudoaneurysm ruptures (ROR: 87.71). Conclusion Our results re-emphasize the importance of a robust Post Marketing Surveillance system and suggest this FAERS database based analysis provides an updated, independent information on Abraxane related AEs to enrich its safety profile. A process of continuous vigilance and additional investigations on specific areas that may have some undesired events are imperative to increase our knowledge on how Abraxane should be handled in terms of its safety.
Collapse
Affiliation(s)
- Yue-Chen Zhao
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xin Li
- Department of Radiology, The Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Chao-Qun Wang
- Department of Gynaecology, Aviation General Hospital, Beijing, People’s Republic of China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Yan-Nan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Tie-Jun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Chao-He Zhang
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
21
|
Jahandoost A, Dashti R, Houshmand M, Hosseini SA. Utilizing machine learning and molecular dynamics for enhanced drug delivery in nanoparticle systems. Sci Rep 2024; 14:26677. [PMID: 39496651 PMCID: PMC11535187 DOI: 10.1038/s41598-024-73268-0] [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: 07/27/2024] [Accepted: 09/16/2024] [Indexed: 11/06/2024] Open
Abstract
Materials data science and machine learning (ML) are pivotal in advancing cancer treatment strategies beyond traditional methods like chemotherapy. Nanotherapeutics, which merge nanotechnology with targeted drug delivery, exemplify this advancement by offering improved precision and reduced side effects in cancer therapy. The development of these nanotherapeutic agents depends critically on understanding nanoparticle (NP) properties and their biological interactions, often analyzed through molecular dynamics (MD) simulations. This study enhances these analyses by integrating ML with MD simulations, significantly improving both prediction accuracy and computational efficiency. We introduce a comprehensive three-stage methodology for predicting the solvent-accessible surface area (SASA) of NPs, which is crucial for their therapeutic efficacy. The process involves training an ML model to forecast the many-body tensor representation (MBTR) for future time steps, applying data augmentation to increase dataset realism, and refining the SASA predictor with both augmented and original data. Results demonstrate that our methodology can predict SASA values 299 time steps ahead with a 40-fold speed improvement and a 25% accuracy increase over existing methods. Importantly, it provides a 300-fold increase in computational speed compared to traditional simulation techniques, offering substantial cost and time savings for nanotherapeutic research and development.
Collapse
Affiliation(s)
- Alireza Jahandoost
- Department of Computer Engineering, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Razieh Dashti
- Department of Nanotechnology, Graduate University of Advanced Technology, Kerman, Iran
| | - Mahboobeh Houshmand
- Department of Computer Engineering, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Seyyed Abed Hosseini
- Department of Electrical Engineering, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
22
|
Shafiei FS, Abroun S. Recent advancements in nanomedicine as a revolutionary approach to treating multiple myeloma. Life Sci 2024; 356:122989. [PMID: 39197575 DOI: 10.1016/j.lfs.2024.122989] [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: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024]
Abstract
Multiple myeloma, the second most common hematological malignancy, remains incurable with a 5-year survival rate of approximately 50 % and recurrence rates near 100 %, despite significant attempts to develop effective medicines. Therefore, there is a pressing demand in the medical field for innovative and more efficient treatments for MM. Currently, the standard approach for treating MM involves administering high-dose chemotherapy, which frequently correlates with improved results; however, one major limiting factor is the significant side effects of these medications. Furthermore, the strategies used to deliver medications to tumors limit their efficacy, whether by rapid clearance from circulation or an insufficient concentration in cancer cells. Cancer treatment has shifted from cytotoxic, nonspecific chemotherapy regimens to molecularly targeted, rationally developed drugs with improved efficacy and fewer side effects. Nanomedicines may provide an effective alternative way to avoid these limits by delivering drugs into the complicated bone marrow microenvironment and efficiently reaching myeloma cells. Putting drugs into nanoparticles can make their pharmacokinetic and pharmacodynamic profiles much better. This can increase the drug's effectiveness in tumors, extend its time in circulation in the blood, and lower its off-target toxicity. In this review, we introduce several criteria for the rational design of nanomedicine to achieve the best anti-tumoral therapeutic results. Next, we discuss recent advances in nanomedicine for MM therapy.
Collapse
Affiliation(s)
- Fatemeh Sadat Shafiei
- Department Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Saeid Abroun
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
23
|
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
|
24
|
Siddique R, Nabi G, Khan S. Editorial: Molecular mechanisms and immunotherapeutic targets in nanomedicine-based cancer therapy. Front Oncol 2024; 14:1468397. [PMID: 39399180 PMCID: PMC11466929 DOI: 10.3389/fonc.2024.1468397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/05/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Rabeea Siddique
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhenzhou, China
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ghulam Nabi
- Department of Zoology, Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore, Pakistan
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhenzhou, China
- First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
25
|
Zhang Y, Tian J. Strategies, Challenges, and Prospects of Nanoparticles in Gynecological Malignancies. ACS OMEGA 2024; 9:37459-37504. [PMID: 39281920 PMCID: PMC11391544 DOI: 10.1021/acsomega.4c04573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Abstract
Gynecologic cancers are a significant health issue for women globally. Early detection and successful treatment of these tumors are crucial for the survival of female patients. Conventional therapies are often ineffective and harsh, particularly in advanced stages, necessitating the exploration of new therapy options. Nanotechnology offers a novel approach to biomedicine. A novel biosensor utilizing bionanotechnology can be employed for early tumor identification and therapy due to the distinctive physical and chemical characteristics of nanoparticles. Nanoparticles have been rapidly applied in the field of gynecologic malignancies, leading to significant advancements in recent years. This study highlights the significance of nanoparticles in treating gynecological cancers. It focuses on using nanoparticles for precise diagnosis and continuous monitoring of the disease, innovative imaging, and analytic methods, as well as multifunctional drug delivery systems and targeted therapies. This review examines several nanocarrier systems, such as dendrimers, liposomes, nanocapsules, and nanomicelles, for gynecological malignancies. The review also examines the enhanced therapeutic potential and targeted delivery of ligand-functionalized nanoformulations for gynecological cancers compared to nonfunctionalized anoformulations. In conclusion, the text also discusses the constraints and future exploration prospects of nanoparticles in chemotherapeutics. Nanotechnology will offer precise methods for diagnosing and treating gynecological cancers.
Collapse
Affiliation(s)
- Yingfeng Zhang
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jing Tian
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| |
Collapse
|
26
|
Chen Y, Zhou Q, Jia Z, Cheng N, Zhang S, Chen W, Wang L. Enhancing cancer immunotherapy: Nanotechnology-mediated immunotherapy overcoming immunosuppression. Acta Pharm Sin B 2024; 14:3834-3854. [PMID: 39309502 PMCID: PMC11413684 DOI: 10.1016/j.apsb.2024.05.032] [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: 03/22/2024] [Revised: 05/12/2024] [Accepted: 05/24/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy is an important cancer treatment method that offers hope for curing cancer patients. While immunotherapy has achieved initial success, a major obstacle to its widespread adoption is the inability to benefit the majority of patients. The success or failure of immunotherapy is closely linked to the tumor's immune microenvironment. Recently, there has been significant attention on strategies to regulate the tumor immune microenvironment in order to stimulate anti-tumor immune responses in cancer immunotherapy. The distinctive physical properties and design flexibility of nanomedicines have been extensively utilized to target immune cells (including tumor-associated macrophages (TAMs), T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated fibroblasts (TAFs)), offering promising advancements in cancer immunotherapy. In this article, we have reviewed treatment strategies aimed at targeting various immune cells to regulate the tumor immune microenvironment. The focus is on cancer immunotherapy models that are based on nanomedicines, with the goal of inducing or enhancing anti-tumor immune responses to improve immunotherapy. It is worth noting that combining cancer immunotherapy with other treatments, such as chemotherapy, radiotherapy, and photodynamic therapy, can maximize the therapeutic effects. Finally, we have identified the challenges that nanotechnology-mediated immunotherapy needs to overcome in order to design more effective nanosystems.
Collapse
Affiliation(s)
- Yunna Chen
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qianqian Zhou
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Zongfang Jia
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Nuo Cheng
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Sheng Zhang
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Weidong Chen
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Lei Wang
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| |
Collapse
|
27
|
Al-Thani AN, Jan AG, Abbas M, Geetha M, Sadasivuni KK. Nanoparticles in cancer theragnostic and drug delivery: A comprehensive review. Life Sci 2024; 352:122899. [PMID: 38992574 DOI: 10.1016/j.lfs.2024.122899] [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: 03/15/2024] [Revised: 06/27/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
This comprehensive review provides an in-depth analysis of how nanotechnology has revolutionized cancer theragnostic, which combines diagnostic and therapeutic methods to customize cancer treatment. The study examines the unique attributes, uses, and difficulties linked to different types of nanoparticles, including gold, iron oxide, silica, Quantum dots, Carbon nanotubes, and liposomes, in the context of cancer treatment. In addition, the paper examines the progression of nanotheranostics, emphasizing its uses in precise medication administration, photothermal therapy, and sophisticated diagnostic methods such as MRI, CT, and fluorescence imaging. Moreover, the article highlights the capacity of nanoparticles to improve the effectiveness of drugs, reduce the overall toxicity in the body, and open up new possibilities for treating cancer by releasing drugs in a controlled manner and targeting specific areas. Furthermore, it tackles concerns regarding the compatibility of nanoparticles and their potential harmful effects, emphasizing the significance of continuous study to improve nanotherapeutic methods for use in medical treatments. The review finishes by outlining potential future applications of nanotechnology in predictive oncology and customized medicine.
Collapse
Affiliation(s)
- Alshayma N Al-Thani
- College of Arts and Sciences, Department of Biological and Environmental Science, Qatar
| | - Asma Ghafoor Jan
- College of Arts and Sciences, Department of Biological and Environmental Science, Qatar
| | - Mohamed Abbas
- Centre for Advanced Materials, Qatar University, Qatar.
| | - Mithra Geetha
- Centre for Advanced Materials, Qatar University, Qatar
| | - Kishor Kumar Sadasivuni
- Centre for Advanced Materials, Qatar University, Qatar; Centre for Advanced Materials, Qatar University, Qatar Department of Mechanical and Industrial Engineering, Qatar
| |
Collapse
|
28
|
Li JH, Hsin PY, Hsiao YC, Chen BJ, Zhuang ZY, Lee CW, Lee WJ, Vo TTT, Tseng CF, Tseng SF, Lee IT. A Narrative Review: Repurposing Metformin as a Potential Therapeutic Agent for Oral Cancer. Cancers (Basel) 2024; 16:3017. [PMID: 39272875 PMCID: PMC11394296 DOI: 10.3390/cancers16173017] [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: 07/22/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Oral cancer, particularly oral squamous cell carcinoma (OSCC), is a significant global health challenge because of its high incidence and limited treatment options. Major risk factors, including tobacco use, alcohol consumption, and specific microbiota, contribute to the disease's prevalence. Recently, a compelling association between diabetes mellitus (DM) and oral cancer has been identified, with metformin, a widely used antidiabetic drug, emerging as a potential therapeutic agent across various cancers, including OSCC. This review explores both preclinical and clinical studies to understand the mechanisms by which metformin may exert its anticancer effects, such as inhibiting cancer cell proliferation, inducing apoptosis, and enhancing the efficacy of existing treatments. Preclinical studies demonstrate that metformin modulates crucial metabolic pathways, reduces inflammation, and impacts cellular proliferation, thereby potentially lowering cancer risk and improving patient outcomes. Additionally, metformin's ability to reverse epithelial-to-mesenchymal transition (EMT), regulate the LIN28/let-7 axis, and its therapeutic role in head and neck squamous cell carcinoma (HNSCC) are examined through experimental models. In clinical contexts, metformin shows promise in enhancing therapeutic outcomes and reducing recurrence rates, although challenges such as drug interactions, complex dosing regimens, and risks such as vitamin B12 deficiency remain. Future research should focus on optimizing metformin's application, investigating its synergistic effects with other therapies, and conducting rigorous clinical trials to validate its efficacy in OSCC treatment. This dual exploration underscores metformin's potential to play a transformative role in both diabetes management and cancer care, potentially revolutionizing oral cancer treatment strategies.
Collapse
Affiliation(s)
- Jui-Hsiang Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - Pei-Yi Hsin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yung-Chia Hsiao
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Bo-Jun Chen
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Zhi-Yun Zhuang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Wei-Ju Lee
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| | - Thi Thuy Tien Vo
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam
| | - Chien-Fu Tseng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei 10048, Taiwan
- Department of Dentistry, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - Shih-Fen Tseng
- Department of Emergency Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
29
|
Jangid H, Singh S, Kashyap P, Singh A, Kumar G. Advancing biomedical applications: an in-depth analysis of silver nanoparticles in antimicrobial, anticancer, and wound healing roles. Front Pharmacol 2024; 15:1438227. [PMID: 39175537 PMCID: PMC11338803 DOI: 10.3389/fphar.2024.1438227] [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: 05/25/2024] [Accepted: 07/26/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction: Silver nanoparticles (AgNPs) have gained significant attention in biomedical applications due to their unique physicochemical properties. This review focuses on the roles of AgNPs in antimicrobial activity, anticancer therapy, and wound healing, highlighting their potential to address critical health challenges. Methods: A bibliometric analysis was conducted using publications from the Scopus database, covering research from 2002 to 2024. The study included keyword frequency, citation patterns, and authorship networks. Data was curated with Zotero and analyzed using Bibliometrix R and VOSviewer for network visualizations. Results: The study revealed an increasing trend in research on AgNPs, particularly in antimicrobial applications, leading to 8,668 publications. Anticancer and wound healing applications followed, with significant contributions from India and China. The analysis showed a growing focus on "green synthesis" methods, highlighting a shift towards sustainable production. Key findings indicated the effectiveness of AgNPs in combating multidrug-resistant bacteria, inducing apoptosis in cancer cells, and promoting tissue regeneration in wound healing. Discussion: The widespread research and applications of AgNPs underscore their versatility in medical interventions. The study emphasizes the need for sustainable synthesis methods and highlights the potential risks, such as long-term toxicity and environmental impacts. Future research should focus on optimizing AgNP formulations for clinical use and further understanding their mechanisms of action. Conclusion: AgNPs play a pivotal role in modern medicine, particularly in addressing antimicrobial resistance, cancer treatment, and wound management. Ongoing research and international collaboration are crucial for advancing the safe and effective use of AgNPs in healthcare.
Collapse
Affiliation(s)
- Himanshu Jangid
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab, India
| | - Sudhakar Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab, India
| | - Piyush Kashyap
- School of Agriculture, Lovely Professional University, Jalandhar, Punjab, India
| | - Avtar Singh
- School of Electrical Engineering and Computing (SoEEC), Adama Science and Technology University (AS-TU), Adama, Ethiopia
| | - Gaurav Kumar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab, India
| |
Collapse
|
30
|
Mariano S, Carata E, Calcagnile L, Panzarini E. Recent Advances in Photodynamic Therapy: Metal-Based Nanoparticles as Tools to Improve Cancer Therapy. Pharmaceutics 2024; 16:932. [PMID: 39065629 PMCID: PMC11280090 DOI: 10.3390/pharmaceutics16070932] [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: 06/17/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer remains a significant global health challenge, with traditional therapies like surgery, chemotherapy, and radiation often accompanied by systemic toxicity and damage to healthy tissues. Despite progress in treatment, these approaches have limitations such as non-specific targeting, systemic toxicity, and resistance development in cancer cells. In recent years, nanotechnology has emerged as a revolutionary frontier in cancer therapy, offering potential solutions to these challenges. Nanoparticles, due to their unique physical and chemical properties, can carry therapeutic payloads, navigate biological barriers, and selectively target cancer cells. Metal-based nanoparticles, in particular, offer unique properties suitable for various therapeutic applications. Recent advancements have focused on the integration of metal-based nanoparticles to enhance the efficacy and precision of photodynamic therapy. Integrating nanotechnology into cancer therapy represents a paradigm shift, enabling the development of strategies with enhanced specificity and reduced off-target effects. This review aims to provide a comprehensive understanding of the pivotal role of metal-based nanoparticles in photodynamic therapy. We explore the mechanisms, biocompatibility, and applications of metal-based nanoparticles in photodynamic therapy, highlighting the challenges and the limitations in their use, as well as the combining of metal-based nanoparticles/photodynamic therapy with other strategies as a synergistic therapeutic approach for cancer treatment.
Collapse
Affiliation(s)
- Stefania Mariano
- Department of Mathematics and Physics, University of Salento, 73100 Lecce, Italy; (S.M.); (L.C.)
| | - Elisabetta Carata
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Lucio Calcagnile
- Department of Mathematics and Physics, University of Salento, 73100 Lecce, Italy; (S.M.); (L.C.)
- CEDAD (CEntre of Applied Physics, DAtation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 73100 Lecce, Italy
| | - Elisa Panzarini
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| |
Collapse
|
31
|
Kong JC, Zhou F, Shi L, Wei Y, Wu C. A novel nanodrug for the sensitization of photothermal chemotherapy for breast cancer in vitro. RSC Adv 2024; 14:21292-21299. [PMID: 38974230 PMCID: PMC11225340 DOI: 10.1039/d4ra01611d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Owing to the complexity of tumor treatment, clinical tumor treatment has evolved from a single treatment mode to multiple combined treatment modes. Reducing the tolerance of tumors to heat and the toxicity of chemotherapy drugs to the body, as well as increasing the sensitivity of tumors to photothermal therapy and chemotherapy drugs, are key issues that urgently need to be addressed in the current cancer treatment. In this work, polylactic acid-based drug nanoparticles (PLA@DOX/GA/ICG) were synthesized with good photothermal conversion ability by encapsulating the water-soluble anticancer drug doxorubicin (DOX), photothermal conversion agent indocyanine green (ICG) and liposoluble drug gambogic acid (GA) using a double emulsion method. The preparation process of PLA@DOX/GA/ICG was examined. Gambogic acid entrapped in PLA@DOX/GA/ICG nanoparticles could act as an HSP90 protein inhibitor to achieve bidirectional sensitization to chemotherapy and photothermal therapy under 808 nm laser irradiation for the first time, effectively ablating breast cancer cells in vitro. This nanodrug was expected to be used for the efficient treatment of tumors.
Collapse
Affiliation(s)
- Ji Chuan Kong
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Feng Zhou
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Liting Shi
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Yihui Wei
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Chunhong Wu
- Henan Polytechinc University Jiaozuo Henan 45400 China
| |
Collapse
|
32
|
Wang D, Li X, Yao H, Liu X, Gao Y, Cong H, Yu B, Shen Y. Hydrophobic modification of polysaccharides and the construction and properties of their micelles: a review of applications in the field of biomedicine. Sci China Chem 2024; 67:1881-1903. [DOI: 10.1007/s11426-023-1916-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/04/2024] [Indexed: 01/06/2025]
|
33
|
Petrovic S, Bita B, Barbinta-Patrascu ME. Nanoformulations in Pharmaceutical and Biomedical Applications: Green Perspectives. Int J Mol Sci 2024; 25:5842. [PMID: 38892030 PMCID: PMC11172476 DOI: 10.3390/ijms25115842] [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: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
This study provides a brief discussion of the major nanopharmaceuticals formulations as well as the impact of nanotechnology on the future of pharmaceuticals. Effective and eco-friendly strategies of biofabrication are also highlighted. Modern approaches to designing pharmaceutical nanoformulations (e.g., 3D printing, Phyto-Nanotechnology, Biomimetics/Bioinspiration, etc.) are outlined. This paper discusses the need to use natural resources for the "green" design of new nanoformulations with therapeutic efficiency. Nanopharmaceuticals research is still in its early stages, and the preparation of nanomaterials must be carefully considered. Therefore, safety and long-term effects of pharmaceutical nanoformulations must not be overlooked. The testing of nanopharmaceuticals represents an essential point in their further applications. Vegetal scaffolds obtained by decellularizing plant leaves represent a valuable, bioinspired model for nanopharmaceutical testing that avoids using animals. Nanoformulations are critical in various fields, especially in pharmacy, medicine, agriculture, and material science, due to their unique properties and advantages over conventional formulations that allows improved solubility, bioavailability, targeted drug delivery, controlled release, and reduced toxicity. Nanopharmaceuticals have transitioned from experimental stages to being a vital component of clinical practice, significantly improving outcomes in medical fields for cancer treatment, infectious diseases, neurological disorders, personalized medicine, and advanced diagnostics. Here are the key points highlighting their importance. The significant challenges, opportunities, and future directions are mentioned in the final section.
Collapse
Affiliation(s)
- Sanja Petrovic
- Department of Chemical Technologies, Faculty of Technology, University of Nis, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia;
| | - Bogdan Bita
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Magurele, Romania;
| | - Marcela-Elisabeta Barbinta-Patrascu
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Magurele, Romania;
| |
Collapse
|
34
|
Garrigós MM, de Oliveira FA, Costa CJS, Rodrigues LR, Nucci MP, Alves ADH, Mamani JB, Rego GNDA, Munoz JM, Gamarra LF. Assessing the toxicity of one-step-synthesized PEG-coated gold nanoparticles: in vitro and in vivo studies. EINSTEIN-SAO PAULO 2024; 22:eAO0764. [PMID: 38775605 PMCID: PMC11081025 DOI: 10.31744/einstein_journal/2024ao0764] [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: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE To evaluate the in vitro and in vivo toxicities of polyethylene glycol-coated gold nanoparticles synthesized using a one-step process. METHODS Gold nanoparticles were prepared via a co-precipitation method using polyethylene glycol, and the synthesis product was characterized. For the in vitro evaluation, a flow cytometry analysis with Annexin V and iodide propidium staining was used to assess cytotoxicity in MG-63 cells labeled with 10, 50, and 100µg/mL of nanoparticle concentration. For the in vivo evaluation, nanoparticles were administered intraperitoneally at a dose of 10mg/kg dose in 10-week-old mice. Toxicity was assessed 24 hours and 7 days after administration via histopathological analysis of various tissues, as well as through renal, hepatic, and hematopoietic evaluations. RESULTS Synthesized nanoparticles exhibited different hydrodynamic sizes depending on the medium: 51.27±1.62nm in water and 268.12±28.45nm (0 hour) in culture medium. They demonstrated a maximum absorbance at 520nm and a zeta potential of -8.419mV. Cellular viability exceeded 90%, with less than 3% early apoptosis, 6% late apoptosis, and 1% necrosis across all labeling conditions, indicating minimal cytotoxicity differences. Histopathological analysis highlighted the accumulation of nanoparticles in the mesentery; however, no lesions or visible agglomeration was observed in the remaining tissues. Renal, hepatic, and hematopoietic analyses showed no significant differences at any time point. CONCLUSION Polyethylene glycol-coated gold nanoparticles exhibit extremely low toxicity and high biocompatibility, showing promise for future studies.
Collapse
Affiliation(s)
- Murilo Montenegro Garrigós
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Cícero Júlio Silva Costa
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Lucas Renan Rodrigues
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Mariana Penteado Nucci
- Hospital das ClínicasFaculdade MedicinaUniversidade de São PauloSão PauloSPBrazil LIM44 - Hospital das Clínicas, Faculdade Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Arielly da Hora Alves
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Javier Bustamante Mamani
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Juan Matheus Munoz
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Lionel Fernel Gamarra
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
35
|
Patel J, Kumar GS, Roy H, Maddiboyina B, Leporatti S, Bohara RA. From nature to nanomedicine: bioengineered metallic nanoparticles bridge the gap for medical applications. DISCOVER NANO 2024; 19:85. [PMID: 38724833 PMCID: PMC11082127 DOI: 10.1186/s11671-024-04021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
The escalating global challenge of antimicrobial resistance demands innovative approaches. This review delves into the current status and future prospects of bioengineered metallic nanoparticles derived from natural sources as potent antimicrobial agents. The unique attributes of metallic nanoparticles and the abundance of natural resources have sparked a burgeoning field of research in combating microbial infections. A systematic review of the literature was conducted, encompassing a wide range of studies investigating the synthesis, characterization, and antimicrobial mechanisms of bioengineered metallic nanoparticles. Databases such as PubMed, Scopus, Web of Science, ScienceDirect, Springer, Taylor & Francis online and OpenAthen were extensively searched to compile a comprehensive overview of the topic. The synthesis methods, including green and sustainable approaches, were examined, as were the diverse biological sources used in nanoparticle fabrication. The amalgamation of metallic nanoparticles and natural products has yielded promising antimicrobial agents. Their multifaceted mechanisms, including membrane disruption, oxidative stress induction, and enzyme inhibition, render them effective against various pathogens, including drug-resistant strains. Moreover, the potential for targeted drug delivery systems using these nanoparticles has opened new avenues for personalized medicine. Bioengineered metallic nanoparticles derived from natural sources represent a dynamic frontier in the battle against microbial infections. The current status of research underscores their remarkable antimicrobial efficacy and multifaceted mechanisms of action. Future prospects are bright, with opportunities for scalability and cost-effectiveness through sustainable synthesis methods. However, addressing toxicity, regulatory hurdles, and environmental considerations remains crucial. In conclusion, this review highlights the evolving landscape of bioengineered metallic nanoparticles, offering valuable insights into their current status and their potential to revolutionize antimicrobial therapy in the future.
Collapse
Affiliation(s)
- Jitendra Patel
- Gitam School of Pharmacy, GITAM (Deemed to be University), Hyderabad Campus, Rudraram, Sangareddy, Hyderabad, TS, 502329, India
| | - G Shiva Kumar
- Gitam School of Pharmacy, GITAM (Deemed to be University), Hyderabad Campus, Rudraram, Sangareddy, Hyderabad, TS, 502329, India
| | - Harekrishna Roy
- Department of Pharmaceutics, Nirmala College of Pharmacy, Mangalagiri, Guntur, Andhra Pradesh, 522503, India.
| | - Balaji Maddiboyina
- Department of Medical and Scientific Communications, Scientific Writing Services, Freyr Global Regulatory Solutions & Services, Phoenix SEZ, Hitech City, Gachibowli, Hyderabad, 500081, India.
| | - Stefano Leporatti
- CNR Nanotec-Istituto Di Nanotecnologia, C\O Campus EcotekneVia Monteroni, 3100, Lecce, Italy
| | - Raghvendra A Bohara
- D.Y. Patil Education Society (Deemed to be University), Kolhapur, MS, India.
- University of Galway, Galway, Ireland.
| |
Collapse
|
36
|
Moghaddam FD, Zare EN, Hassanpour M, Bertani FR, Serajian A, Ziaei SF, Paiva-Santos AC, Neisiany RE, Makvandi P, Iravani S, Xu Y. Chitosan-based nanosystems for cancer diagnosis and therapy: Stimuli-responsive, immune response, and clinical studies. Carbohydr Polym 2024; 330:121839. [PMID: 38368115 DOI: 10.1016/j.carbpol.2024.121839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 02/19/2024]
Abstract
Cancer, a global health challenge of utmost severity, necessitates innovative approaches beyond conventional treatments (e.g., surgery, chemotherapy, and radiation therapy). Unfortunately, these approaches frequently fail to achieve comprehensive cancer control, characterized by inefficacy, non-specific drug distribution, and the emergence of adverse side effects. Nanoscale systems based on natural polymers like chitosan have garnered significant attention as promising platforms for cancer diagnosis and therapy owing to chitosan's inherent biocompatibility, biodegradability, nontoxicity, and ease of functionalization. Herein, recent advancements pertaining to the applications of chitosan nanoparticles in cancer imaging and drug/gene delivery are deliberated. The readers are introduced to conventional non-stimuli-responsive and stimuli-responsive chitosan-based nanoplatforms. External triggers like light, heat, and ultrasound and internal stimuli such as pH and redox gradients are highlighted. The utilization of chitosan nanomaterials as contrast agents or scaffolds for multimodal imaging techniques e.g., magnetic resonance, fluorescence, and nuclear imaging is represented. Key applications in targeted chemotherapy, combination therapy, photothermal therapy, and nucleic acid delivery using chitosan nanoformulations are explored for cancer treatment. The immunomodulatory effects of chitosan and its role in impacting the tumor microenvironment are analyzed. Finally, challenges, prospects, and future outlooks regarding the use of chitosan-based nanosystems are discussed.
Collapse
Affiliation(s)
- Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133 Rome, Italy
| | | | - Mahnaz Hassanpour
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Francesca Romana Bertani
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133 Rome, Italy
| | - Azam Serajian
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Seyedeh Farnaz Ziaei
- Department of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ana Cláudia Paiva-Santos
- Drug Development and Technology Laboratory, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Rasoul Esmaeely Neisiany
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland; Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran.
| | - Pooyan Makvandi
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, UK; The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 324000 Quzhou, Zhejiang, China; Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India; Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| |
Collapse
|
37
|
Ranasinghe R, Mathai M, Abdullah Alshawsh M, Zulli A. Nanocarrier-mediated cancer therapy with cisplatin: A meta-analysis with a promising new paradigm. Heliyon 2024; 10:e28171. [PMID: 39839154 PMCID: PMC11747978 DOI: 10.1016/j.heliyon.2024.e28171] [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: 08/25/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 01/23/2025] Open
Abstract
Aims Cisplatin is a frontline chemotherapeutic utilized to attenuate multiple cancers in the clinic. Given its side-effects, a new cisplatin formulation which could prevent cytotoxicity, metabolic deficiencies and metastasis is much needed. This study investigates whether nanocarriers can provide a better mode of drug delivery in preclinical cancer models seeking a potent anticancer therapeutic agent. Materials and methods The PubMed database was searched, and 242 research articles were screened from which 94 articles qualified for selection from those published by December 31, 2023 and the data was synthesized using the Review Manager software. Key findings Cisplatin encapsulated as a nanomedicine confirmed the versatility of nanocarriers in significantly diminishing cancer cell viability, half maximal inhibitory concentration, tumour volume, biodistribution of platinum in tumours and kidney; at p < 0.00001 and a 95% confidence interval. Significance An estimated 19.3 million global cancer incidence is reported with 50% mortality worldwide for which nanocarrier-mediated cisplatin therapy is most promising. Our findings offer new vistas for future cancer treatment when combined with chemo-immunotherapy that utilizes the recently advanced nanozymes.
Collapse
Affiliation(s)
- Ranmali Ranasinghe
- Institute for Health and Sport, College of Health and Medicine, Victoria University, Melbourne, Victoria, Australia
| | - Michael Mathai
- Institute for Health and Sport, College of Health and Medicine, Victoria University, Melbourne, Victoria, Australia
| | - Mohammed Abdullah Alshawsh
- Department of Paediatrics, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Anthony Zulli
- Institute for Health and Sport, College of Health and Medicine, Victoria University, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Pavone AM, Benfante V, Giaccone P, Stefano A, Torrisi F, Russo V, Serafini D, Richiusa S, Pometti M, Scopelliti F, Ippolito M, Giannone AG, Cabibi D, Asti M, Vettorato E, Morselli L, Merone M, Lunardon M, Andrighetto A, Tuttolomondo A, Cammarata FP, Verona M, Marzaro G, Mastrotto F, Parenti R, Russo G, Comelli A. Biodistribution Assessment of a Novel 68Ga-Labeled Radiopharmaceutical in a Cancer Overexpressing CCK2R Mouse Model: Conventional and Radiomics Methods for Analysis. Life (Basel) 2024; 14:409. [PMID: 38541733 PMCID: PMC10972008 DOI: 10.3390/life14030409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 01/12/2025] Open
Abstract
The aim of the present study consists of the evaluation of the biodistribution of a novel 68Ga-labeled radiopharmaceutical, [68Ga]Ga-NODAGA-Z360, injected into Balb/c nude mice through histopathological analysis on bioptic samples and radiomics analysis of positron emission tomography/computed tomography (PET/CT) images. The 68Ga-labeled radiopharmaceutical was designed to specifically bind to the cholecystokinin receptor (CCK2R). This receptor, naturally present in healthy tissues such as the stomach, is a biomarker for numerous tumors when overexpressed. In this experiment, Balb/c nude mice were xenografted with a human epidermoid carcinoma A431 cell line (A431 WT) and overexpressing CCK2R (A431 CCK2R+), while controls received a wild-type cell line. PET images were processed, segmented after atlas-based co-registration and, consequently, 112 radiomics features were extracted for each investigated organ / tissue. To confirm the histopathology at the tissue level and correlate it with the degree of PET uptake, the studies were supported by digital pathology. As a result of the analyses, the differences in radiomics features in different body districts confirmed the correct targeting of the radiopharmaceutical. In preclinical imaging, the methodology confirms the importance of a decision-support system based on artificial intelligence algorithms for the assessment of radiopharmaceutical biodistribution.
Collapse
Affiliation(s)
- Anna Maria Pavone
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.M.P.); (V.R.); (R.P.)
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (V.B.); (P.G.); (A.C.)
| | - Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (V.B.); (P.G.); (A.C.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Paolo Giaccone
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (V.B.); (P.G.); (A.C.)
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy; (S.R.); (F.P.C.); (G.R.)
- Laboratori Nazionali del Sud, National Institute for Nuclear Physics, INFN-LNS, 95123 Catania, Italy
| | - Filippo Torrisi
- Medicine and Surgery Department, University of Enna “Kore”, 94019 Enna, Italy;
| | - Vincenzo Russo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.M.P.); (V.R.); (R.P.)
| | - Davide Serafini
- Legnaro National Laboratories, Italian Institute of Nuclear Physics, Viale Dell’Università 2, 35020 Padova, Italy; (D.S.); (L.M.); (A.A.)
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Selene Richiusa
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy; (S.R.); (F.P.C.); (G.R.)
| | - Marco Pometti
- Nuclear Medicine Department, Cannizzaro Hospital, 95126 Catania, Italy; (M.P.); (F.S.); (M.I.)
| | - Fabrizio Scopelliti
- Nuclear Medicine Department, Cannizzaro Hospital, 95126 Catania, Italy; (M.P.); (F.S.); (M.I.)
| | - Massimo Ippolito
- Nuclear Medicine Department, Cannizzaro Hospital, 95126 Catania, Italy; (M.P.); (F.S.); (M.I.)
| | - Antonino Giulio Giannone
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.G.G.); (D.C.)
| | - Daniela Cabibi
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.G.G.); (D.C.)
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42122 Reggio Emilia, Italy;
| | - Elisa Vettorato
- Department of Pharmaceutical Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| | - Luca Morselli
- Legnaro National Laboratories, Italian Institute of Nuclear Physics, Viale Dell’Università 2, 35020 Padova, Italy; (D.S.); (L.M.); (A.A.)
| | - Mario Merone
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Marcello Lunardon
- Department of Physics and Astronomy, University of Padova, Via Marzolo 8, 35131 Padova, Italy;
| | - Alberto Andrighetto
- Legnaro National Laboratories, Italian Institute of Nuclear Physics, Viale Dell’Università 2, 35020 Padova, Italy; (D.S.); (L.M.); (A.A.)
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Francesco Paolo Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy; (S.R.); (F.P.C.); (G.R.)
- Laboratori Nazionali del Sud, National Institute for Nuclear Physics, INFN-LNS, 95123 Catania, Italy
| | - Marco Verona
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (M.V.); (G.M.); (F.M.)
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (M.V.); (G.M.); (F.M.)
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (M.V.); (G.M.); (F.M.)
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.M.P.); (V.R.); (R.P.)
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy; (S.R.); (F.P.C.); (G.R.)
- Laboratori Nazionali del Sud, National Institute for Nuclear Physics, INFN-LNS, 95123 Catania, Italy
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (V.B.); (P.G.); (A.C.)
| |
Collapse
|
39
|
Lahmadi S, Alamery S, Beagan A, Alotaibi K, Alswieleh A. Advanced hybrid silica nanoparticles with pH-responsive diblock copolymer brushes: optimized design for controlled doxorubicin loading and release in cancer therapy. RSC Adv 2024; 14:8819-8828. [PMID: 38495996 PMCID: PMC10941263 DOI: 10.1039/d4ra00282b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024] Open
Abstract
This study delves into the development, characterization, and application of modified mesoporous silica nanoparticles (MSNs) for targeted drug delivery in cancer therapy. MSNs were functionalized with poly(2-(diisopropylamino)ethyl methacrylate) (PDPA) and poly(glycidyl methacrylate) (PGMA), and further modified with cross-linkers DAE and Ornithine. Characterization using FT-IR, SEM, TEM, DLS, and XPS confirmed the successful surface modifications, revealing particle sizes primarily within the 63-94 nm range. The MSNs demonstrated a pH-responsive behavior, crucial for smart drug delivery. Loading and release studies using Doxorubicin (DOX) showed a controlled release, with an 8 μg mg-1 loading capacity. Cytotoxicity assays on Caco2 colon cancer cells revealed that unloaded nano-systems, at concentrations above 45 μM, resulted in approximately 60% cell death, indicating inherent anti-cancer properties. However, variations in cytotoxic effects were observed in drug-loaded MSNs, with some modifications showing reduced anti-cancer activity. These findings highlight the potential of MSNs in drug delivery and cancer treatment, emphasizing the importance of nanoparticle design in therapeutic efficacy.
Collapse
Affiliation(s)
- Shatha Lahmadi
- Department of Chemistry, College of Science, King Saud University Riyadh Kingdom of Saudi Arabia
| | - Salman Alamery
- Department of Biochemistry, College of Science, King Saud University Riyadh Kingdom of Saudi Arabia
| | - Abeer Beagan
- Department of Chemistry, College of Science, King Saud University Riyadh Kingdom of Saudi Arabia
| | - Khalid Alotaibi
- Department of Chemistry, College of Science, King Saud University Riyadh Kingdom of Saudi Arabia
| | - Abdullah Alswieleh
- Department of Chemistry, College of Science, King Saud University Riyadh Kingdom of Saudi Arabia
| |
Collapse
|
40
|
Wang B, Xu XJ, Fu Y, Ren B, Yang XD, Yang HY. A tumor-targeted and enzyme-responsive gold nanorod-based nanoplatform with facilitated endo-lysosomal escape for synergetic photothermal therapy and protein therapy. Dalton Trans 2024; 53:2120-2130. [PMID: 38180436 DOI: 10.1039/d3dt03305h] [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: 01/06/2024]
Abstract
To tackle the obstacles related to tumor targeting and overcome the limitations of single treatment models, we have developed a nanoplatform that is both tumor-targeted and enzyme-responsive. This nanoplatform integrates photothermal gold nanorods (AuNRs) and protein drugs into a single system. This nanosystem, known as AuNRs@HA-mPEG-Deta-LA, was fabricated by modifying gold nanorods (AuNRs) with a polymeric ligand called hyaluronic acid-grafted-(mPEG/diethylenetriamine-conjugated-lipoic acid). The purpose of this fabrication was to load cytochrome c (CC) and utilize it for the synergetic protein-photothermal therapy of cancer. The resulting nanoplatform exhibited a high efficiency in loading proteins and demonstrated excellent stability in different biological environments. Additionally, CC-loaded AuNRs@HA-mPEG-Deta-LA not only enabled localized hyperthermia for photothermal therapy (PTT) with laser irradiation but also facilitated the release of CC under the action of hyaluronidase, an enzyme known to be overexpressed in tumor cells. The confocal imaging results demonstrated that the presence of a specific polymeric ligand on this nanoparticle enhances the internalization of CD44-positive cancer cells, accelerates endo/lysosomal escape, and facilitates the controlled release of CC within the cells. Furthermore, the results of the MTT assay also showed that AuNRs@HA-mPEG-Deta-LA as a protein nanocarrier demonstrated excellent biocompatibility. Importantly, this synergistic therapeutic strategy effectively induced apoptosis in A549 cancer cells by increasing the intracellular concentration of CC and utilizing the photothermal conversion of AuNRs, which was observed to be more effective compared to using only protein therapy or PTT. Therefore, this study showcased a nanoplatform based on AuNRs that has great potential for tumor-targeted protein delivery in combination with PTT in cancer treatment.
Collapse
Affiliation(s)
- Bo Wang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Xin Jun Xu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Bo Ren
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Xiao Dong Yang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| |
Collapse
|
41
|
Rajan SS, Chandran R, Abrahamse H. Overcoming challenges in cancer treatment: Nano-enabled photodynamic therapy as a viable solution. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1942. [PMID: 38456341 DOI: 10.1002/wnan.1942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Cancer presents a formidable challenge, necessitating innovative therapies that maximize effectiveness while minimizing harm to healthy tissues. Nanotechnology has emerged as a transformative force in cancer treatment, particularly through nano-enabled photodynamic therapy (NE-PDT), which leverages precise and targeted interventions. NE-PDT capitalizes on photosensitizers activated by light to generate reactive oxygen species (ROS) that initiate apoptotic pathways in cancer cells. Nanoparticle enhancements optimize this process, improving drug delivery, selectivity, and ROS production within tumors. This review dissects NE-PDT's mechanistic framework, showcasing its potential to harness apoptosis as a potent tool in cancer therapy. Furthermore, the review explores the synergy between NE-PDT and complementary treatments like chemotherapy, immunotherapy, and targeted therapies, highlighting the potential to amplify apoptotic responses, enhance immune recognition of cancer cells, and inhibit resistance mechanisms. Preclinical and clinical advancements in NE-PDT demonstrate its efficacy across various cancer types. Challenges in translating NE-PDT into clinical practice are also addressed, emphasizing the need for optimizing nanoparticle design, refining dosimetry, and ensuring long-term safety. Ultimately, NE-PDT represents a promising approach in cancer therapy, utilizing the intricate mechanisms of apoptosis to address therapeutic hurdles. The review underscores the importance of understanding the interplay between nanoparticles, ROS generation, and apoptotic pathways, contributing to a deeper comprehension of cancer biology and novel therapeutic strategies. As interdisciplinary collaborations continue to thrive, NE-PDT offers hope for effective and targeted cancer interventions, where apoptosis manipulation becomes central to conquering cancer. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Sheeja S Rajan
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
42
|
Quiñonero G, Gallo J, Carrasco A, Samitier J, Villasante A. Engineering Biomimetic Nanoparticles through Extracellular Vesicle Coating in Cancer Tissue Models. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3097. [PMID: 38132993 PMCID: PMC10746063 DOI: 10.3390/nano13243097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Using nanoparticles (NPs) in drug delivery has exhibited promising therapeutic potential in various cancer types. Nevertheless, several challenges must be addressed, including the formation of the protein corona, reduced targeting efficiency and specificity, potential immune responses, and issues related to NP penetration and distribution within 3-dimensional tissues. To tackle these challenges, we have successfully integrated iron oxide nanoparticles into neuroblastoma-derived extracellular vesicles (EVs) using the parental labeling method. We first developed a tissue-engineered (TE) neuroblastoma model, confirming the viability and proliferation of neuroblastoma cells for at least 12 days, supporting its utility for EV isolation. Importantly, EVs from long-term cultures exhibited no differences compared to short-term cultures. Concurrently, we designed Rhodamine (Rh) and Polyacrylic acid (PAA)-functionalized magnetite nanoparticles (Fe3O4@PAA-Rh) with high crystallinity, purity, and superparamagnetic properties (average size: 9.2 ± 2.5 nm). We then investigated the internalization of Fe3O4@PAA-Rh nanoparticles within neuroblastoma cells within the TE model. Maximum accumulation was observed overnight while ensuring robust cell viability. However, nanoparticle internalization was low. Taking advantage of the enhanced glucose metabolism exhibited by cancer cells, glucose (Glc)-functionalized nanoparticles (Fe3O4@PAA-Rh-Glc) were synthesized, showing superior cell uptake within the 3D model without inducing toxicity. These glucose-modified nanoparticles were selected for parental labeling of the TE models, showing effective NP encapsulation into EVs. Our research introduces innovative approaches to advance NP delivery, by partially addressing the challenges associated with 3D systems, optimizing internalization, and enhancing NP stability and specificity through EV-based carriers. Also, our findings hold the promise of more precise and effective cancer therapies while minimizing potential side effects.
Collapse
Affiliation(s)
- Gema Quiñonero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Juan Gallo
- Advanced Magnetic Theranostic Nanostructures Laboratory, International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal
| | - Alex Carrasco
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Electronic and Biomedical Engineering, University of Barcelona, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Aranzazu Villasante
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Electronic and Biomedical Engineering, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
43
|
Wang B, Zhang Y, Yin X. Advances in tumor immunomodulation based on nanodrug delivery systems. Front Immunol 2023; 14:1297493. [PMID: 38106403 PMCID: PMC10725201 DOI: 10.3389/fimmu.2023.1297493] [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: 09/20/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Immunotherapy is a therapeutic approach that employs immunological principles and techniques to enhance and amplify the body's immune response, thereby eradicating tumor cells. Immunotherapy has demonstrated effective antitumor effects on a variety of malignant tumors. However, when applied to humans, many immunotherapy drugs fail to target lesions with precision, leading to an array of adverse immune-related reactions that profoundly limit the clinical application of immunotherapy. Nanodrug delivery systems enable the precise delivery of immunotherapeutic drugs to targeted tissues or specific immune cells, enhancing the immune antitumor effect while reducing the number of adverse reactions. A nanodrug delivery system provides a feasible strategy for activating the antitumor immune response by the following mechanisms: 1) increased targeting and uptake of vaccines by DCs, which enhances the efficacy of the immune response; 2) increased tumor cell immunogenicity; 3) regulation of TAMs and other cells by, for example, regulating the polarization of TAMs and interfering with TAN formation, and ECM remodeling by CAFs; and 4) interference with tumor immune escape signaling pathways, namely, the PD-1/PD-L1, FGL1/LAG-3 and IDO signaling pathways. This paper reviews the progress of nanodrug delivery system research with respect to tumor immunotherapy based on tumor immunomodulation over the last few years, discussing the promising future of these delivery systems under this domain.
Collapse
Affiliation(s)
- Bo Wang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, China
| | - Yue Zhang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, China
| | - Xunzhe Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
44
|
Fernandes Q, Therachiyil L, Khan AQ, Bedhiafi T, Korashy HM, Bhat AA, Uddin S. Shrinking the battlefield in cancer therapy: Nanotechnology against cancer stem cells. Eur J Pharm Sci 2023; 191:106586. [PMID: 37729956 DOI: 10.1016/j.ejps.2023.106586] [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: 06/24/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
Cancer remains one of the leading causes of mortality worldwide, presenting a significant healthcare challenge owing to the limited efficacy of current treatments. The application of nanotechnology in cancer treatment leverages the unique optical, magnetic, and electrical attributes of nanomaterials to engineer innovative, targeted therapies. Specifically, manipulating nanomaterials allows for enhanced drug loading efficiency, improved bioavailability, and targeted delivery systems, reducing the non-specific cytotoxic effects characteristic of conventional chemotherapies. Furthermore, recent advances in nanotechnology have demonstrated encouraging results in specifically targeting CSCs, a key development considering the role of these cells in disease recurrence and resistance to treatment. Despite these breakthroughs, the clinical approval rates of nano-drugs have not kept pace with research advances, pointing to existing obstacles that must be addressed. In conclusion, nanotechnology presents a novel, powerful tool in the fight against cancer, particularly in targeting the elusive and treatment-resistant CSCs. This comprehensive review delves into the intricacies of nanotherapy, explicitly targeting cancer stem cells, their markers, and associated signaling pathways.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, Qatar University, Doha, Qatar; Translational Cancer Research Facility, Hamad Medical Corporation, National Center for Cancer Care and Research, PO. Box 3050, Doha, Qatar
| | - Lubna Therachiyil
- Academic Health System, Hamad Medical Corporation, Translational Research Institute, Doha 3050, Qatar; Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Abdul Q Khan
- Academic Health System, Hamad Medical Corporation, Translational Research Institute, Doha 3050, Qatar
| | - Takwa Bedhiafi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahab Uddin
- College of Medicine, Qatar University, Doha, Qatar; Academic Health System, Hamad Medical Corporation, Dermatology Institute, Doha 3050, Qatar; Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 22602, India.
| |
Collapse
|
45
|
Abdoul-Latif FM, Ainane A, Houmed Aboubaker I, Mohamed J, Ainane T. An Overview of Cancer in Djibouti: Current Status, Therapeutic Approaches, and Promising Endeavors in Local Essential Oil Treatment. Pharmaceuticals (Basel) 2023; 16:1617. [PMID: 38004482 PMCID: PMC10674319 DOI: 10.3390/ph16111617] [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: 10/07/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Djibouti, a developing economy, grapples with significant socioeconomic obstacles and the prevalence of infectious pathologies, including certain forms of neoplasms. These challenges are exacerbated by limited access to affordable medical technologies for diagnosis, coupled with a lack of preventive interventions, particularly in disadvantaged areas. The attention devoted to local phytotherapeutic treatments underscores the uniqueness of Djibouti's flora, resulting from its distinctive geographical position. International focus specifically centers on harnessing this potential as a valuable resource, emphasizing the phytoconstituents used to counter pathologies, notably carcinomas. This comprehensive overview covers a broad spectrum, commencing with an examination of the current state of knowledge, namely an in-depth investigation of oncological risk factors. Essential elements of control are subsequently studied, highlighting the fundamental prerequisites for effective management. The significance of dietary habits in cancer prevention and support is explored in depth, while traditional methods are examined, highlighting the cultural significance of indigenous essential oil therapies and encouraging further research based on the promising results.
Collapse
Affiliation(s)
- Fatouma Mohamed Abdoul-Latif
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti P.O. Box 486, Djibouti;
| | - Ayoub Ainane
- Superior School of Technology of Khenifra (EST-Khenifra), University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| | | | - Jalludin Mohamed
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti P.O. Box 486, Djibouti;
| | - Tarik Ainane
- Superior School of Technology of Khenifra (EST-Khenifra), University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| |
Collapse
|
46
|
Younis MK, Khalil IA, Younis NS, Fakhr Eldeen RR, Abdelnaby RM, Aldeeb RA, Taha AA, Hassan DH. Aceclofenac/Citronellol Oil Nanoemulsion Repurposing Study: Formulation, In Vitro Characterization, and In Silico Evaluation of Their Antiproliferative and Pro-Apoptotic Activity against Melanoma Cell Line. Biomedicines 2023; 11:2531. [PMID: 37760972 PMCID: PMC10525854 DOI: 10.3390/biomedicines11092531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Aceclofenac (ACF) is a widely used non-steroidal anti-inflammatory drug (NSAID) known for its effectiveness in treating pain and inflammation. Recent studies have demonstrated that ACF possesses antiproliferative properties, inhibiting the growth of cancer cells in various cancer cell lines. Citronellol, a monoterpenoid alcohol found in essential oils, exhibits antioxidant properties and activities such as inhibiting cell growth and acetylcholinesterase inhibition. In this study, the objective was to formulate and evaluate an aceclofenac/citronellol oil nanoemulsion for its antiproliferative effects on melanoma. The optimal concentrations of citronellol oil, Tween 80, and Transcutol HP were determined using a pseudoternary phase diagram. The formulated nanoemulsions were characterized for droplet size, zeta potential, thermophysical stability, and in vitro release. The selected formula (F1) consisted of citronellol oil (1 gm%), Tween 80 (4 gm%), and Transcutol HP (1 gm%). F1 exhibited a spherical appearance with high drug content, small droplet size, and acceptable negative zeta potential. The amorphous state of the drug in the nanoemulsion was confirmed by Differential Scanning Calorimetry, while FTIR analysis indicated its homogenous solubility. The nanoemulsion showed significant antiproliferative activity, with a lower IC50 value compared to aceclofenac or citronellol alone. Flow cytometric analysis revealed cell cycle arrest and increased apoptosis induced by the nanoemulsion. In silico studies provided insights into the molecular mechanism underlying the observed antitumor activity. In conclusion, the developed aceclofenac/citronellol oil nanoemulsion exhibited potent cytotoxicity and pro-apoptotic effects, suggesting its potential as a repurposed antiproliferative agent for melanoma treatment. In a future plan, further animal model research for validation is suggested.
Collapse
Affiliation(s)
- Mona K. Younis
- Department of Pharmaceutics, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12566, Egypt; (I.A.K.); (R.A.A.); (A.A.T.); (D.H.H.)
| | - Islam A. Khalil
- Department of Pharmaceutics, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12566, Egypt; (I.A.K.); (R.A.A.); (A.A.T.); (D.H.H.)
| | - Nancy S. Younis
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Zagazig University Hospitals, Zagazig 44519, Egypt
| | - Rasha R. Fakhr Eldeen
- Department of Biochemistry, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12566, Egypt;
| | - Rana M. Abdelnaby
- Department Pharmaceutical Chemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Reem A. Aldeeb
- Department of Pharmaceutics, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12566, Egypt; (I.A.K.); (R.A.A.); (A.A.T.); (D.H.H.)
| | - Amal A. Taha
- Department of Pharmaceutics, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12566, Egypt; (I.A.K.); (R.A.A.); (A.A.T.); (D.H.H.)
| | - Doaa H. Hassan
- Department of Pharmaceutics, College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12566, Egypt; (I.A.K.); (R.A.A.); (A.A.T.); (D.H.H.)
| |
Collapse
|
47
|
Mishra I, Garrett M, Curry S, Jameson J, Kastellorizios M. Effect of Composition and Size on Surface Properties of Anti-Cancer Nanoparticles. Int J Mol Sci 2023; 24:13417. [PMID: 37686222 PMCID: PMC10487715 DOI: 10.3390/ijms241713417] [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: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
Liposomal formulations offer significant advantages as anticancer drug carriers for targeted drug delivery; however, due to their complexity, clinical translation has been challenging. In addition, liposomal product manufacturing has been interrupted in the past, as was the case for Doxil® (doxorubicin hydrochloride liposome injection). Here, interfacial tension (IFT) measurements were investigated as a potential physicochemical characterization tool to aid in liposomal product characterization during development and manufacturing. A pendant drop method using an optical tensiometer was used to measure the interfacial tension of various analogues of Doxil® liposomal suspensions in air and in dodecane. The effect of liposome concentration, formulation (PEG and cholesterol content), presence of encapsulated drug, as well as average particle size was analyzed. It was observed that Doxil® analog liposomes demonstrate surfactant-like behavior with a sigmoidal-shape interfacial tension vs. concentration curve. This behavior was heavily dependent on PEG content, with a complete loss of surfactant-like behavior when PEG was removed from the formulation. In addition to interfacial tension, three data analyses were identified as able to distinguish between formulations with variations in PEG, cholesterol, and particle size: (i) polar and non-polar contribution to interfacial tension, (ii) liposomal concentration at which the polar and non-polar components were equal, and (iii) rate of interfacial tension decay after droplet formation, which is indicative of how quickly liposomes migrate from the bulk of the solution to the surface. We demonstrate for the first time that interfacial tension can be used to detect certain liposomal formulation changes, such as PEG content, encapsulated drug presence, and size variability, and may make a useful addition to physicochemical characterization during development and manufacturing of liposomal products.
Collapse
Affiliation(s)
| | | | | | | | - Michail Kastellorizios
- Department of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|