1
|
Alkatheeri A, Salih S, Kamil N, Alnuaimi S, Abuzar M, Abdelrahman SS. Nano-Radiopharmaceuticals in Colon Cancer: Current Applications, Challenges, and Future Directions. Pharmaceuticals (Basel) 2025; 18:257. [PMID: 40006069 PMCID: PMC11859487 DOI: 10.3390/ph18020257] [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: 12/24/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Colon cancer remains a significant global health challenge; however, the treatment outcome for colon patients can be improved through early detection and effective treatment. Nano-radiopharmaceuticals, combining nanotechnology with radiopharmaceuticals, are emerging as a revolutionary approach in both colon cancer diagnostic imaging and therapy, playing a significant role in the management of colon cancer patients. This review examines the use of nano-radiopharmaceuticals in the diagnosis and treatment of colon cancer, highlighting current applications, challenges, and future directions. Nanocarriers of radionuclides have shown potential in improving cancer treatment, including liposomes, microparticles, nanoparticles, micelles, dendrimers, and hydrogels, which are approved by the FDA. These nanocarriers can deliver targeted drugs into malignant cells without affecting normal cells, reducing side effects. Antibody-guided systemic radionuclide-targeted therapy has shown potential for treating cancer. Novel cancer nanomedicines, like Hensify and 32P BioSilicon, are under clinical development for targeted radiation delivery in percutaneous intratumoral injections. Although using nano-radiopharmaceuticals is a superior technique for diagnosing and treating colon cancer, there are limitations and challenges, such as the unintentional accumulation of nanoparticles in healthy tissues, which leads to toxicity due to biodistribution issues, as well as high manufacturing costs that limit their availability for patients. However, the future direction is moving toward providing more precise radiopharmaceuticals, which is crucial for enhancing the diagnosis and treatment of colon cancer and reducing production costs.
Collapse
Affiliation(s)
- Ajnas Alkatheeri
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates;
| | - Suliman Salih
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates;
- National Cancer Institute, University of Gezira, Wad Madani 2667, Sudan
| | - Noon Kamil
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | - Sara Alnuaimi
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | - Memona Abuzar
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | | |
Collapse
|
2
|
Liu H, Lu Y, Zong J, Zhang B, Li X, Qi H, Yu T, Li Y. Engineering dendritic cell biomimetic membrane as a delivery system for tumor targeted therapy. J Nanobiotechnology 2024; 22:663. [PMID: 39465376 PMCID: PMC11520105 DOI: 10.1186/s12951-024-02913-7] [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: 05/17/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024] Open
Abstract
Targeted immunotherapies make substantial strides in clinical cancer care due to their ability to counteract the tumor's capacity to suppress immune responses. Advances in biomimetic technology with minimally immunogenic and highly targeted, are addressing issues of targeted drug delivery and disrupting the tumor's immunosuppressive environment to trigger immune activation. Specifically, the use of dendritic cell (DC) membranes to coat nanoparticles ensures targeted delivery due to DC's unique ability to activate naive T cells, spotlighting their role in immunotherapy aimed at disrupting the tumor microenvironment. The potential of DC's biomimetic membrane to mediate immune activation and target tumors is gaining momentum, enhancing the effectiveness of cancer treatments in conjunction with other immune responses. This review delves into the methodologies behind crafting DC membranes and the fusion of dendritic and tumor cell membranes for encapsulating therapeutic nanoparticles. It explores their applications and recent advancements in combating cancer, offering an all-encompassing perspective on DC biomimetic nanosystems, immunotherapy driven by antigen presentation, and the collaborative efforts of drug delivery in chemotherapy and photodynamic therapies. Current evidence shows promise in augmenting combined therapeutic approaches for cancer treatment and holds translational potential for various cancer treatments in a clinical setting.
Collapse
Affiliation(s)
- Huiyang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Yiming Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China.
| |
Collapse
|
3
|
Al-Nasser S, Abdulla MH, Alhassan N, Vaali-Mohammed MA, Al-Omar S, Hamdi N, Elnakady Y, Matou-Nasri S, Mansour L. A Benzimidazole-Based N-Heterocyclic Carbene Derivative Exhibits Potent Antiproliferative and Apoptotic Effects against Colorectal Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1379. [PMID: 39336420 PMCID: PMC11433580 DOI: 10.3390/medicina60091379] [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: 05/01/2024] [Revised: 06/20/2024] [Accepted: 08/05/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: Colorectal cancer (CRC) remains a major global health issue. Although chemotherapy is the first-line treatment, its effectiveness is limited due to drug resistance developed in CRC. To overcome resistance and improve the prognosis of CRC patients, investigating new therapeutic approaches is necessary. Materials and Methods: Using human colorectal adenocarcinoma (HT29) and metastatic CRC (SW620) cell lines, the potential anticancer properties of a newly synthesized compound 1-(Isobutyl)-3-(4-methylbenzyl) benzimidazolium chloride (IMBZC) were evaluated by performing MTT cytotoxicity, cell migration, and colony formation assays, as well as by monitoring apoptosis-related protein and gene expression using Western blot and reverse transcription-quantitative polymerase chain reaction technologies. Results: Tested at various concentrations, the half-maximal inhibitory concentrations (IC50) of IMBZC on HT29 and SW620 cell growth were determined to be 22.13 µM (6.97 μg/mL) and 15.53 µM (4.89 μg/mL), respectively. IMBZC did not alter the cell growth of normal HEK293 cell lines. In addition, IMBZC inhibited cell migration and significantly decreased colony formation, suggesting its promising role in suppressing cancer metastasis. Mechanistic analyses revealed that IMBZC treatment increased the expression of pro-apoptotic proteins p53 and Bax, while decreasing the expression of anti-apoptotic proteins Bcl-2 and Bcl-xL, thus indicating the induction of apoptosis in IMBZC-treated CRC cells, compared to untreated cells. Additionally, the addition of IMBZC to conventional chemotherapeutic drugs (i.e., 5-fluorouracil, irinotecan, and oxaliplatin) resulted in an increase in the cytotoxic potential of the drugs. Conclusions: This study suggests that IMBZC has substantial anticancer effects against CRC cells through its ability to induce apoptosis, inhibit cancer cell migration and colony formation, and enhance the cytotoxic effects of conventional chemotherapeutic drugs. These findings indicate that IMBZC could be a promising chemotherapeutic drug for the treatment of CRC. Further research should be conducted using in vivo models to confirm the anti-CRC activities of IMBZC.
Collapse
Affiliation(s)
- Sarah Al-Nasser
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Noura Alhassan
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Mansoor-Ali Vaali-Mohammed
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Suliman Al-Omar
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naceur Hamdi
- Research Laboratory of Environmental Sciences and Technologies (LR16ES09), Higher Institute of Environmental Sciences and Technology, University of Carthage, Hammam-Lif 2050, Tunisia
| | - Yasser Elnakady
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School of Systems Biology, George Mason University, Manassas, VA 20110, USA
| | - Lamjed Mansour
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
4
|
Xu P, Zhong S, Wei Y, Duan X, Zhang M, Shen W, Ma Y, Zhang YH. Surface-Functionalized Halo-Tag Gold Nanoprobes for Live-Cell Long-Term Super-Resolution Imaging of Endoplasmic Reticulum Dynamics. ACS NANO 2024; 18:21433-21446. [PMID: 39080511 DOI: 10.1021/acsnano.4c06138] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Super-resolution fluorescence microscopy has emerged as a powerful tool for studying endoplasmic reticulum (ER) dynamics in living cells. However, the lack of high-brightness, high-photostability, and stable labeling probes makes long-term super-resolution imaging of the ER still challenging. Herein, we reported a surface-functionalized Halo-tag gold nanofluorescent probe (GNP-Atto565-fR8-CA) that exhibits excellent brightness, photostability, and biocompatibility. GNP-Atto565-fR8-CA can simultaneously load multiple Atto565 dye molecules, significantly improving its brightness. Modifying the cell-penetrating peptide fR8 enables GNP-Atto565-fR8-CA to be efficiently delivered into the cytoplasm, overcoming the challenge of their easy entrapment in vesicles. Fluorescent labeling of ER proteins via Halo tags enables high specificity and stable labeling of GNP-Atto565-fR8-CA to the ER. The SIM super-resolution imaging results showed that GNP-Atto565-fR8-CA can track and observe the long-term dynamic process of the ER, and can also be used for long-term super-resolution imaging of the dynamic interactions between the ER and other organelles. This work offers a practical tool to study live-cell ER ultrastructure and dynamics.
Collapse
Affiliation(s)
- Peng Xu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Wuhan Forth Hospital, Wuhan, Hubei 430030, China
| | - Simei Zhong
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yunfei Wei
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xinxin Duan
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Meng Zhang
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wei Shen
- Wuhan Forth Hospital, Wuhan, Hubei 430030, China
| | - Yan Ma
- Joint Wuhan Blood Center-Huazhong University of Science and Technology Hematology Optical Imaging Center, Institute of Blood Transfusion of Hubei Province, Wuhan Blood Center, Wuhan, Hubei 430030, China
| | - Yu-Hui Zhang
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
5
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
6
|
Shi Y, Sun Y, Shen X, Yang Z, Xu B, Bao C. Combination of handgrip strength and high-sensitivity modified Glasgow prognostic score predicts survival outcomes in patients with colon cancer. Front Nutr 2024; 11:1421560. [PMID: 39010859 PMCID: PMC11247022 DOI: 10.3389/fnut.2024.1421560] [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: 04/22/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024] Open
Abstract
Objective Handgrip strength (HGS) and the high-sensitivity modified Glasgow prognostic score (HS-mGPS) are associated with the survival of patients with cancer. However, no studies have investigated the combined effect of HGS and HS-mGPS on the overall survival (OS) of patients with colon cancer. Methods Prospective follow-up data of colon cancer patients undergoing radical resection from April, 2016 to September, 2019 were retrospectively collected. We combined the HGS and HS-mGPS to create a new composite index, HGS-HS-mGPS. The hazard ratio (HR) and 95% confidence interval (CI) were calculated using Cox regression models to assess the association between variables and OS. Risk factors on OS rates were investigated by Cox analyses and the nomogram was constructed using significant predictors and HGS-HS-mGPS. The predictive performance of the nomogram was evaluated by receiver operating characteristic curve and calibration curve. Results This study included a total of 811 patients, of which 446 (55.0%) were male. The HGS optimal cut-off values of male and female patients were 28.8 and 19.72 kg, respectively. Multivariate analysis revealed that low HGS and high HS-mGPS were independent risk factors of colon cancer after adjusting confounders (adjusted HR = 3.20; 95% CI: 2.27-4.50; p < 0.001 and adjusted HR = 1.55; 95% CI: 1.12-2.14; p = 0.008 respectively). Patients with low HGS and high HS-mGPS had a 10.76-fold higher mortality risk than those with neither (adjusted HR = 10.76; 95% CI: 5.38-21.54; p < 0.001). A nomogram predicting 1-, 3-, and 5 year OS was constructed based on three clinicopathologic prognostic factors. Importantly, incorporating HGS-HS-mGPS into the nomogram model meaningfully improved the predictive performance. The decision curve analyses demonstrated the application value of the HGS-HS-mGPS nomogram for predicting OS of patients with colon cancer. Conclusion HGS-HS-mGPS is associated with the survival of patients with colon cancer. These findings indicate the usefulness of HGS and HS-mGPS measurements in clinical practice for improving patient assessment, cancer prognosis, and precise intervention.
Collapse
Affiliation(s)
- Yifan Shi
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuting Sun
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Xiaoming Shen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Zenghui Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Binghua Xu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chuanqing Bao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
Yin T, Han J, Cui Y, Shang D, Xiang H. Prospect of Gold Nanoparticles in Pancreatic Cancer. Pharmaceutics 2024; 16:806. [PMID: 38931925 PMCID: PMC11207630 DOI: 10.3390/pharmaceutics16060806] [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: 01/11/2024] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 06/28/2024] Open
Abstract
Pancreatic cancer (PC) is characterized by its notably poor prognosis and high mortality rate, underscoring the critical need for advancements in its diagnosis and therapy. Gold nanoparticles (AuNPs), with their distinctive physicochemical characteristics, demonstrate significant application potential in cancer therapy. For example, upon exposure to lasers of certain wavelengths, they facilitate localized heating, rendering them extremely effective in photothermal therapy. Additionally, their extensive surface area enables the conjugation of therapeutic agents or targeting molecules, increasing the accuracy of drug delivery systems. Moreover, AuNPs can serve as radiosensitizers, enhancing the efficacy of radiotherapy by boosting the radiation absorption in tumor cells. Here, we systematically reviewed the application and future directions of AuNPs in the diagnosis and treatment of PC. Although AuNPs have advantages in improving diagnostic and therapeutic efficacy, as well as minimizing damage to normal tissues, concerns about their potential toxicity and safety need to be comprehensively evaluated.
Collapse
Affiliation(s)
- Tianyi Yin
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (T.Y.); (J.H.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Jingrun Han
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (T.Y.); (J.H.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Yuying Cui
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (T.Y.); (J.H.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Hong Xiang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| |
Collapse
|
8
|
Ahmadpour MR, Yousefi M, Rakhshandeh H, Darroudi M, Mousavi SH, Soukhtanloo M, Sabouri Z, Askari VR, Hashemzadeh A, Manjiri MA, Motavasselian M. Biosynthesis of Gold Nanoparticles Using Quince Seed Water Extract and Investigation of Their Anticancer Effect Against Cancer Cell Lines. IEEE Trans Nanobioscience 2024; 23:118-126. [PMID: 37379200 DOI: 10.1109/tnb.2023.3287805] [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: 06/30/2023]
Abstract
In this study, gold nanoparticles (Au-NPs) were synthesized using HAuCl4 and quince seed mucilage (QSM) extract, which was characterized by conventional methods including Fourier transforms electron microscopy (FTIR), UV-Visible spectroscopy (UV-Vis), Field emission electron microscopy (FESEM), Transmission electron microscopy (TEM), Dynamic light spectroscopy (DLS), and Zeta-potential. The QSM acted as reductant and stabilizing agents simultaneously. The NP's anticancer activity was also investigated against osteosarcoma cell lines (MG-63), which showed an IC50 of [Formula: see text]/mL.
Collapse
|
9
|
Ibrahim IAA, Alzahrani AR, Alanazi IM, Shahzad N, Shahid I, Falemban AH, Azlina MFN, Arulselvan P. Carbohydrate polymers-based surface modified nano delivery systems for enhanced target delivery to colon cancer - A review. Int J Biol Macromol 2023; 253:126581. [PMID: 37652322 DOI: 10.1016/j.ijbiomac.2023.126581] [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: 05/29/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Carbohydrate polymers-based surface-modified nano-delivery systems have gained significant attention in recent years for enhancing targeted delivery to colon cancer. These systems leverage carbohydrate polymers' unique properties, such as biocompatibility, biodegradability, and controlled release. These properties make them suitable candidates for drug delivery applications. Nano-delivery systems loaded with bioactive compounds are well-studied for targeted colorectal cancer delivery. However, those drugs' target reach is still limited in various nano-delivery systems. To overcome this limitation, surface modification of nanoparticles with carbohydrate polymers like chitosan, pectin, alginate, and guar gum showed enhanced target-reaching capacity along with enhanced anticancer efficacy. Recently, a chitosan-decorated PLGA nanoparticle was constructed with tannic acid and vitamin E and showed long-term release of specific targets along with higher anticancer efficacy. Similarly, Chitosan-conjugated glucuronic acid-coated silica nanoparticles loaded with capecitabine were studied against colon cancer and found to be the pH-responsive controlled release of capecitabine with higher anticancer efficacy. Surface-modified carbohydrate polymers have promising potential for improving colon cancer target delivery. By leveraging the unique properties of these polymers, such as surface modification, pH responsiveness, mucoadhesion, controlled drug release, and combination therapy, researchers are working toward developing more effective and targeted treatment strategies for colon cancer.
Collapse
Affiliation(s)
- Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ibrahim M Alanazi
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naiyer Shahzad
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Shahid
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Alaa Hisham Falemban
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohd Fahami Nur Azlina
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Malaysia
| | - Palanisamy Arulselvan
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, 602 105, India
| |
Collapse
|
10
|
Roshani M, Rezaian-Isfahni A, Lotfalizadeh MH, Khassafi N, Abadi MHJN, Nejati M. Metal nanoparticles as a potential technique for the diagnosis and treatment of gastrointestinal cancer: a comprehensive review. Cancer Cell Int 2023; 23:280. [PMID: 37981671 PMCID: PMC10657605 DOI: 10.1186/s12935-023-03115-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/27/2023] [Indexed: 11/21/2023] Open
Abstract
Gastrointestinal (GI) cancer is a major health problem worldwide, and current diagnostic and therapeutic approaches are often inadequate. Various metallic nanoparticles (MNPs) have been widely studied for several biomedical applications, including cancer. They may potentially overcome the challenges associated with conventional chemotherapy and significantly impact the overall survival of GI cancer patients. Functionalized MNPs with targeted ligands provide more efficient localization of tumor energy deposition, better solubility and stability, and specific targeting properties. In addition to enhanced therapeutic efficacy, MNPs are also a diagnostic tool for molecular imaging of malignant lesions, enabling non-invasive imaging or detection of tumor-specific or tumor-associated antigens. MNP-based therapeutic systems enable simultaneous stability and solubility of encapsulated drugs and regulate the delivery of therapeutic agents directly to tumor cells, which improves therapeutic efficacy and minimizes drug toxicity and leakage into normal cells. However, metal nanoparticles have been shown to have a cytotoxic effect on cells in vitro. This can be a concern when using metal nanoparticles for cancer treatment, as they may also kill healthy cells in addition to cancer cells. In this review, we provide an overview of the current state of the field, including preparation methods of MNPs, clinical applications, and advances in their use in targeted GI cancer therapy, as well as the advantages and limitations of using metal nanoparticles for the diagnosis and treatment of gastrointestinal cancer such as potential toxicity. We also discuss potential future directions and areas for further research, including the development of novel MNP-based approaches and the optimization of existing approaches.
Collapse
Affiliation(s)
- Mohammad Roshani
- Internal Medicine and Gastroenterology, Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arya Rezaian-Isfahni
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Negar Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hassan Jafari Najaf Abadi
- Research Center for Health Technology Assessment and Medical Informatics, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
11
|
de Paula MC, Carvalho SG, Silvestre ALP, Dos Santos AM, Meneguin AB, Chorilli M. The role of hyaluronic acid in the design and functionalization of nanoparticles for the treatment of colorectal cancer. Carbohydr Polym 2023; 320:121257. [PMID: 37659830 DOI: 10.1016/j.carbpol.2023.121257] [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: 05/22/2023] [Revised: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 09/04/2023]
Abstract
Despite advances in new approaches for colorectal cancer (CRC) therapy, intravenous chemotherapy remains one of the main treatment options; however, it has limitations associated with off-target toxicity, tumor cell resistance due to molecular complexity and CRC heterogeneity, which lead to tumor recurrence and metastasis. In oncology, nanoparticle-based strategies have been designed to avoid systemic toxicity and increase drug accumulation at tumor sites. Hyaluronic acid (HA) has obtained significant attention thanks to its ability to target nanoparticles (NPs) to CRC cells through binding to cluster-determinant-44 (CD44) and hyaluronan-mediated motility (RHAMM) receptors, along with its efficient biological properties of mucoadhesion. This review proposes to discuss the state of the art in HA-based nanoparticulate systems intended for localized treatment of CRC, highlighting the importance of the mucoadhesion and active targeting provided by this polymer. In addition, an overview of CRC will be provided, emphasizing the importance of CD44 and RHAMM receptors in this type of cancer and the current challenges related to this disease, and important concepts about the physicochemical and biological properties of HA will also be addressed. Finally, this review aims to contribute to the advancement of accuracy treatment of CRC by the design of new platforms based on by HA.
Collapse
Affiliation(s)
- Mariana Carlomagno de Paula
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Suzana Gonçalves Carvalho
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Amanda Letícia Polli Silvestre
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Aline Martins Dos Santos
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Andréia Bagliotti Meneguin
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| |
Collapse
|
12
|
Djermane R, Nieto C, Vega MA, Del Valle EMM. Antibody-Loaded Nanoplatforms for Colorectal Cancer Diagnosis and Treatment: An Update. Pharmaceutics 2023; 15:pharmaceutics15051514. [PMID: 37242756 DOI: 10.3390/pharmaceutics15051514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
At present, colorectal cancer (CRC) is the second deadliest type of cancer, partly because a high percentage of cases are diagnosed at advanced stages when tumors have already metastasized. Thus, there is an urgent need to develop novel diagnostic systems that allow early detection as well as new therapeutic systems that are more specific than those currently available. In this context, nanotechnology plays a very important role in the development of targeted platforms. In recent decades, many types of nanomaterials with advantageous properties have been used for nano-oncology applications and have been loaded with different types of targeted agents, capable of recognizing tumor cells or biomarkers. Indeed, among the different types of targeted agents, the most widely used are monoclonal antibodies, as the administration of many of them is already approved by the main drug regulatory agencies for the treatment of several types of cancer, including CRC. In this way, this review comprehensively discusses the main drawbacks of the conventional screening technologies and treatment for CRC, and it presents recent advances in the application of antibody-loaded nanoplatforms for CRC detection, therapy or theranostics applications.
Collapse
Affiliation(s)
- Rania Djermane
- Chemical Engineering Department, University of Salamanca, Plaza de los Caídos s/n, 37008 Salamanca, Spain
| | - Celia Nieto
- Chemical Engineering Department, University of Salamanca, Plaza de los Caídos s/n, 37008 Salamanca, Spain
- Biomedical Research Institute of Salamanca (IBSAL), University Care Complex of Salamanca, Paseo de San Vicente 58, 37007 Salamanca, Spain
| | - Milena A Vega
- Chemical Engineering Department, University of Salamanca, Plaza de los Caídos s/n, 37008 Salamanca, Spain
- Biomedical Research Institute of Salamanca (IBSAL), University Care Complex of Salamanca, Paseo de San Vicente 58, 37007 Salamanca, Spain
| | - Eva M Martín Del Valle
- Chemical Engineering Department, University of Salamanca, Plaza de los Caídos s/n, 37008 Salamanca, Spain
- Biomedical Research Institute of Salamanca (IBSAL), University Care Complex of Salamanca, Paseo de San Vicente 58, 37007 Salamanca, Spain
| |
Collapse
|
13
|
Simón M, Jørgensen JT, Norregaard K, Henriksen JR, Clergeaud G, Andresen TL, Hansen AE, Kjaer A. Neoadjuvant Gold Nanoshell-Based Photothermal Therapy Combined with Liposomal Doxorubicin in a Mouse Model of Colorectal Cancer. Int J Nanomedicine 2023; 18:829-841. [PMID: 36824412 PMCID: PMC9942687 DOI: 10.2147/ijn.s389260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/23/2022] [Indexed: 02/18/2023] Open
Abstract
Introduction Traditional cancer treatments, such as chemotherapy, are often incapable of achieving complete responses as standalone therapies. Hence, current treatment strategies typically rely on a combination of several approaches. Nanoparticle-based photothermal therapy (PTT) is a technique used to kill cancer cells through localized, severe hyperthermia that has shown promise as an add-on treatment to multiple cancer therapies. Here, we evaluated whether the combination of gold nanoshell (NS)-based PTT and liposomal doxorubicin could improve outcome in a mouse model of colorectal cancer. Methods First, NS-based PTT was performed on tumor-bearing mice. Radiolabeled liposomes were then injected at different timepoints to follow their accumulation in the tumor and determine the ideal injection time after PTT. In addition, fluorescent liposomes were used to observe the liposomal distribution in the tumor after PTT. Finally, we combined PTT and doxorubicin-loaded liposomes and studied the effect of the treatment strategy on the mice by following tumor growth and survival. Results PTT significantly improved liposomal accumulation in the tumor, but only when the liposomes were injected immediately after the therapy. The liposomes accumulated mostly in regions adjacent to the ablated areas. When PTT was combined with liposomal doxorubicin, the mice experienced a slowdown in tumor growth and an improvement in survival. Conclusion According to our preclinical study, NS-based PTT seems promising as an add-on treatment for liposomal chemotherapy and potentially other systemic therapies, and could be relevant for future application in a clinical setting.
Collapse
Affiliation(s)
- Marina Simón
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kamilla Norregaard
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Rosager Henriksen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gael Clergeaud
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anders Elias Hansen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark,Correspondence: Andreas Kjaer, Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark, Email
| |
Collapse
|
14
|
Freitas SC, Sanderson D, Caspani S, Magalhães R, Cortés-Llanos B, Granja A, Reis S, Belo JH, Azevedo J, Gómez-Gaviro MV, de Sousa CT. New Frontiers in Colorectal Cancer Treatment Combining Nanotechnology with Photo- and Radiotherapy. Cancers (Basel) 2023; 15:383. [PMID: 36672333 PMCID: PMC9856291 DOI: 10.3390/cancers15020383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
Colorectal cancer is the third most common cancer worldwide. Despite recent advances in the treatment of this pathology, which include a personalized approach using radio- and chemotherapies in combination with advanced surgical techniques, it is imperative to enhance the performance of these treatments and decrease their detrimental side effects on patients' health. Nanomedicine is likely the pathway towards solving this challenge by enhancing both the therapeutic and diagnostic capabilities. In particular, plasmonic nanoparticles show remarkable potential due to their dual therapeutic functionalities as photothermal therapy agents and as radiosensitizers in radiotherapy. Their dual functionality, high biocompatibility, easy functionalization, and targeting capabilities make them potential agents for inducing efficient cancer cell death with minimal side effects. This review aims to identify the main challenges in the diagnosis and treatment of colorectal cancer. The heterogeneous nature of this cancer is also discussed from a single-cell point of view. The most relevant works in photo- and radiotherapy using nanotechnology-based therapies for colorectal cancer are addressed, ranging from in vitro studies (2D and 3D cell cultures) to in vivo studies and clinical trials. Although the results using nanoparticles as a photo- and radiosensitizers in photo- and radiotherapy are promising, preliminary studies showed that the possibility of combining both therapies must be explored to improve the treatment efficiency.
Collapse
Affiliation(s)
- Sara C. Freitas
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Sanderson
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Doctor Esquerdo 46, 28007 Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| | - Sofia Caspani
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Ricardo Magalhães
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | | | - Andreia Granja
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - João Horta Belo
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - José Azevedo
- Colorectal Surgery—Champalimaud Foundation, Champalimaud Centre for the Unknown, Avenida Brasília, 1400-038 Lisboa, Portugal
| | - Maria Victoria Gómez-Gaviro
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Doctor Esquerdo 46, 28007 Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| | - Célia Tavares de Sousa
- IFIMUP-Institute of Physics for Advanced Materials, Nanotechnology and Photonics of University of Porto, LaPMET-Laboratory of Physics for Materials and Emergent Technologies, Departamento de Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
- Departamento de Física Aplicada, Facultad de Ciencias, Universidad Autonoma de Madrid (UAM), Campus de Cantoblanco, C/ Francisco Tomas y Valiente, 7, 28049 Madrid, Spain
| |
Collapse
|
15
|
de Freitas JVB, Reis AVF, Silva ADO, de Sousa ACC, Martins JRP, Nogueira KAB, da Silva Moreira T, Petrilli R, Eloy JO. Monoclonal Antibodies in Nanosystems as a Strategy for Cancer Treatment. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
16
|
Huang X, Kaneda-Nakashima K, Kadonaga Y, Kabayama K, Shimoyama A, Ooe K, Kato H, Toyoshima A, Shinohara A, Haba H, Wang Y, Fukase K. Astatine-211-Labeled Gold Nanoparticles for Targeted Alpha-Particle Therapy via Intravenous Injection. Pharmaceutics 2022; 14:pharmaceutics14122705. [PMID: 36559199 PMCID: PMC9782038 DOI: 10.3390/pharmaceutics14122705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Alpha-particle radiotherapy has gained considerable attention owing to its potent anti-cancer effect. 211At, with a relatively short half-life of 7.2 h, emits an alpha particle within a few cell diameters with high kinetic energy, which damages cancer cells with high biological effectiveness. In this study, we investigated the intravenous injection of 211At-labeled gold nanoparticles (AuNPs) for targeted alpha-particle therapy (TAT). Different kinds of surface-modified gold nanoparticles can be labeled with 211At in high radiochemical yield in 5 min, and no purification is necessary. The in vivo biodistribution results showed the accumulation of 5 nm 211At-AuNPs@mPEG at 2.25% injection dose per gram (% ID/g) in tumors within 3 h via the enhanced permeability and retention (EPR) effect. Additionally, we observed a long retention time in tumor tissues within 24 h. This is the first study to demonstrate the anti-tumor efficacy of 5 nm 211At-AuNPs@mPEG that can significantly suppress tumor growth in a pancreatic cancer model via intravenous administration. AuNPs are satisfactory carriers for 211At delivery, due to simple and efficient synthesis processes and high stability. The intravenous administration of 5 nm 211At-AuNPs@mPEG has a significant anti-tumor effect. This study provides a new framework for designing nanoparticles suitable for targeted alpha-particle therapy via intravenous injection.
Collapse
Affiliation(s)
- Xuhao Huang
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Kazuko Kaneda-Nakashima
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Yuichiro Kadonaga
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Atsushi Shimoyama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Kazuhiro Ooe
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Hiroki Kato
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Atsushi Toyoshima
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Atsushi Shinohara
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Faculty of Health Science, Osaka Aoyama University, 2-11-1 Niina, Minoh 562-8580, Osaka, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako 351-0198, Saitama, Japan
| | - Yang Wang
- Nishina Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako 351-0198, Saitama, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Correspondence: ; Tel.: +81-6-6850-5391
| |
Collapse
|
17
|
Huang J, Wei F, Cui Y, Hou L, Lin T. Fluorescence immunosensor based on functional nanomaterials and its application in tumor biomarker detection. RSC Adv 2022; 12:31369-31379. [PMID: 36349017 PMCID: PMC9624183 DOI: 10.1039/d2ra04989a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2022] [Indexed: 09/29/2023] Open
Abstract
An immunosensor is defined as an analytical device that detects the binding of an antigen to its specific antibody by coupling an immunochemical reaction to the surface of a device called a transducer. Fluorescence immunosensing is one of the most promising immunoassays at present, and has the advantages of simple operation, fast response and high stability. A traditional fluorescence immunosensor often uses an enzyme-labelled antibody as a recognition unit and an organic dye as a fluorescence probe, so it is easily affected by environmental factors with low sensitivity. Nanomaterials have unique photostability, catalytic properties and biocompatibility, which open up a new path for the construction of stable and sensitive fluorescence immunosensors. This paper briefly introduces different kinds of immunosensors and the role of nanomaterials in the construction of immunosensors. The significance of fluorescent immunosensors constructed from functional nanomaterials to detect tumor biomarkers was analyzed, and the strategies to further improve the performance of fluorescent immunosensors and their future development trend were summarized.
Collapse
Affiliation(s)
- Juanjuan Huang
- School of Chemistry and Pharmaceutical Science, State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University Guilin 541004 P. R. China
| | - Fenghuang Wei
- School of Chemistry and Pharmaceutical Science, State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University Guilin 541004 P. R. China
| | - Yuling Cui
- Jinan Center for Food and Drug Control Jinan 250102 Shandong China
| | - Li Hou
- School of Chemistry and Pharmaceutical Science, State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University Guilin 541004 P. R. China
| | - Tianran Lin
- School of Chemistry and Pharmaceutical Science, State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University Guilin 541004 P. R. China
| |
Collapse
|
18
|
Soto KM, Luzardo-Ocampo I, López-Romero JM, Mendoza S, Loarca-Piña G, Rivera-Muñoz EM, Manzano-Ramírez A. Gold Nanoparticles Synthesized with Common Mullein (Verbascum thapsus) and Castor Bean (Ricinus communis) Ethanolic Extracts Displayed Antiproliferative Effects and induced Caspase 3 Activity in Human HT29 and SW480 Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14102069. [PMID: 36297503 PMCID: PMC9609588 DOI: 10.3390/pharmaceutics14102069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 12/19/2022] Open
Abstract
Gold nanoparticles (AuNPs) are promising nanomaterials exhibiting anti-cancer effects. Green AuNPs synthesis using plant extracts can be used to achieve stable and beneficial nanoparticles due to their content of bioactive compounds. This research aimed to synthesize and evaluate the antiproliferative and caspase-3 activity induction of green AuNPs synthesized with common mullein (V. thapsus) flowers (AuNPsME) and castor bean (R. communis) leaves (AuNPsCE) ethanolic extracts in human HT29 and SW480 colorectal cancer cells. Their effect was compared with chemically synthesized AuNPs (AuNPsCS). The extracts mainly contained p-coumaric acid (71.88–79.93 µg/g), ferulic acid (19.07–310.71 µg/g), and rutin (8.14–13.31 µg/g). The obtained nanoparticles presented typical FT-IR bands confirming the inclusion of polyphenols from V. thapsus and R. communis and spherical/quasi-spherical morphologies with diameters in the 20.06–37.14 nm range. The nanoparticles (20–200 µg/mL) showed antiproliferative effects in both cell lines, with AuNPsCE being the most potent (IC50 HT29: 110.10 and IC50SW480: 64.57 µg/mL). The AuNPsCS showed the lowest intracellular reactive oxygen species (ROS) generation in SW480 cells. All treatments induced caspase 3/7 activity to a similar or greater extent than 30 mM H2O2-treated cells. Results indicated the suitability of V. thapsus and R. communis extracts to synthesize AuNPs, displaying a stronger antiproliferative effect than AuNPsCS.
Collapse
Affiliation(s)
- Karen M. Soto
- Centro de Investigaciones y de Estudios Avanzados del I. P. N. Unidad Querétaro, Queretaro 76230, Mexico
- Correspondence: (K.M.S.); (A.M.-R.)
| | - Ivan Luzardo-Ocampo
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM-Campus Juriquilla), Queretaro 76230, Mexico
| | - José M. López-Romero
- Centro de Investigaciones y de Estudios Avanzados del I. P. N. Unidad Querétaro, Queretaro 76230, Mexico
| | - Sandra Mendoza
- Research and Graduate Program in Food Science, Universidad Autónoma de Querétaro, Queretaro 76010, Mexico
| | - Guadalupe Loarca-Piña
- Research and Graduate Program in Food Science, Universidad Autónoma de Querétaro, Queretaro 76010, Mexico
| | - Eric M. Rivera-Muñoz
- Centro de Física Aplicada y Tecnología Avanzada (CFATA), Universidad Nacional Autónoma de México (UNAM-Campus Juriquilla), Queretaro 76230, Mexico
| | - Alejandro Manzano-Ramírez
- Centro de Investigaciones y de Estudios Avanzados del I. P. N. Unidad Querétaro, Queretaro 76230, Mexico
- Correspondence: (K.M.S.); (A.M.-R.)
| |
Collapse
|
19
|
Xie H, Wei L, Yuan G, Liu M, Liang Y, Gao S, Wang Q, Lin X, Tang S, Gan J. Combination of Geriatric Nutritional Risk Index and Carcinoembryonic Antigen to Predict the Survival of Patients With Colorectal Cancer. Front Nutr 2022; 9:902080. [PMID: 35845803 PMCID: PMC9280638 DOI: 10.3389/fnut.2022.902080] [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: 03/22/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background This study explored the value of the combination of Geriatric Nutritional Risk Index (GNRI) and carcinoembryonic antigen (CEA) for the prognosis assessment of CRC patients. Methods This study retrospectively enrolled 1,014 CRC patients who underwent surgery between 2012 and 2014. Kaplan-Meier and log-rank tests were used to compare survival differences. Cox proportional hazards regression analysis was used to assess risk factors associated with progression-free survival (PFS) and overall survival (OS). Nomograms were constructed to predict the prognosis of CRC patients. Randomized internal validation was used to confirm the predictive accuracy of the prognostic nomograms. Results The GNRI-CEA score was established by combining GNRI and CEA. Compared with patients with normal GNRI-CEA scores, patients with mild/moderate/severe GNRI-CEA scores had significantly lower survival (PFS, 68.99% vs. 57.75% vs. 41.34% vs. 31.36%, p < 0.001; OS, 68.99% vs. 57.75% vs. 41.34% vs. 31.36%, p < 0.001). The GNRI-CEA score is an independent factor predicting the prognosis of CRC patients. The risk of death was twofold higher in patients with low GNRI and high CEA than in those with both normal GNRI and CEA [PFS, hazard ratio (HR), 2.339; 95% confidence interval (CI), 1.656-3.303; p < 0.001; OS, HR, 2.340; 95% CI, 1.645-3.329; p < 0.001]. Prognostic nomograms had good resolution and accuracy in predicting 1-5 year PFS and OS. Randomized internal validation showed that the nomograms were reliable. Conclusion The combination of GNRI and CEA can effectively stratify the prognosis of CRC patients. The nomogram established based on the two indices can provide a personalized reference for prognostic assessment and clinical decision-making for CRC patients.
Collapse
Affiliation(s)
- Hailun Xie
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Lishuang Wei
- Department of Geriatric Respiratory Disease Ward, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Guanghui Yuan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Mingxiang Liu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Yanren Liang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Shunhui Gao
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Qiwen Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Xin Lin
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Grade 2018, Department of Clinical Medicine, Guangxi Medical University, Nanning, China
| | - Shuangyi Tang
- Department of Pharmacy, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Jialiang Gan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| |
Collapse
|