1
|
Cammarata A, Marino J, Atia MN, Durán H, Glisoni RJ. Novel doxycycline gold nanoparticles via green synthesis using PEO-PPO block copolymers for enhanced radiosensitization of melanoma. Biomater Sci 2025. [PMID: 40261332 DOI: 10.1039/d5bm00253b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
This study focuses on a green and sustainable nanoplatform for the delivery of therapeutic agents, based on gold nanoparticles (AuNPs) synthesized using PEO-PPO block copolymers (F127, F68, P85, and their F127:P85 combination) as dual-function reducing and stabilizing agents. This eco-friendly approach eliminates the need for toxic chemical reductants, adheres to green chemistry principles, and yields highly stable, biocompatible nanosystems. The resulting polymer-stabilized AuNPs were associated with doxycycline (DOXY), a mitochondrial biogenesis inhibitor with radiosensitizing properties, and characterized using UV-Vis spectroscopy, dynamic light scattering (DLS), transmission electron microscopy (TEM), and X-ray fluorescence (XRF). The nanoparticles exhibited high colloidal stability, with tunable hydrodynamic diameters modulated by the copolymer composition. In vitro studies on A-375 and IIB-MEL-J melanoma cell lines revealed that DOXY-associated AuNPs, combined with gamma radiation (2 Gy, 137Cs), significantly enhanced radiosensitivity, reducing both cell viability and clonogenic survival. The physicochemical features of the nanosystems, particularly particle size and surface composition, influenced cellular uptake and therapeutic response. Notably, AuNPs stabilized with F127:P85 copolymer combination (∼19 nm) outperformed those with F127 (∼30 nm), despite displaying slightly higher polydispersity. Compared to Turkevich AuNPs, our copolymer-coated nanosystems demonstrated superior colloidal stability and cellular internalization. These findings highlight the potential of green-synthesized AuNPs as multifunctional, biocompatible platforms for therapeutic delivery, supporting the development of effective and environmentally responsible multimodal cancer therapies. Moreover, the simplicity, scalability, and cost-effectiveness of the synthesis process support its potential for future translational applications.
Collapse
Affiliation(s)
- Agostina Cammarata
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Biotecnología, Junín 956, C1113AAD Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Nanobiotecnología (NANOBIOTEC), Buenos Aires, Argentina.
| | - Julieta Marino
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Mariel N Atia
- Comisión Nacional de Energía Atómica (CNEA), Gerencia de Investigación y Aplicaciones, Subgerencia de Tecnología y Aplicaciones de Aceleradores, San Martín, Buenos Aires, Argentina
- Instituto de Nanociencia y Nanotecnología (INN-CNEA-CONICET), San Martín, Buenos Aires, Argentina
| | - Hebe Durán
- Comisión Nacional de Energía Atómica (CNEA), Gerencia de Investigación y Aplicaciones, Subgerencia de Tecnología y Aplicaciones de Aceleradores, San Martín, Buenos Aires, Argentina
- Instituto de Nanociencia y Nanotecnología (INN-CNEA-CONICET), San Martín, Buenos Aires, Argentina
- Universidad Nacional de San Martín, Escuela de Ciencia y Tecnología, San Martín, Buenos Aires, Argentina
| | - Romina J Glisoni
- Universidad de Buenos Aires (UBA), Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Biotecnología, Junín 956, C1113AAD Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Nanobiotecnología (NANOBIOTEC), Buenos Aires, Argentina.
| |
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. [PMID: 40191672 PMCID: PMC11670031 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
|
Li D, Wang J, Li X, Wang Z, Yu Q, Koh SB, Wu R, Ye L, Guo Y, Okoli U, Pati-Alam A, Mota E, Wei W, Yoo KH, Cho WC, Feng D, Heavey S. Interactions between radiotherapy resistance mechanisms and the tumor microenvironment. Crit Rev Oncol Hematol 2025; 210:104705. [PMID: 40107436 DOI: 10.1016/j.critrevonc.2025.104705] [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: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Resistance to radiotherapy (RT) presents a significant clinical challenge in management of cancer. Recent evidence points to specific mechanisms of resistance within the tumor microenvironment (TME), which we aim to discuss, with the aim of overcoming the clinical challenge. METHODS We performed the narrative review using PubMed and Web of Science databases to identify studies that reported the regulative network and treatments of RT resistance from TME perspectives. RESULTS RT significantly changes the immune TME of cancers, which is closely appearing to play a key role in RT resistance (RTR) by modulating immune cell infiltration and function. Various phenotypes are involved in the development of RTR, such as autophagy, senescence, oxidative stress, cell polarization, ceramide metabolism, and angiogenesis in the TME. Key genes and pathways are also implicated in RTR, including immune and inflammatory cytokines, TGF-β, P53, the NF-κB pathway, the cGAS/STING pathway, the ERK and AKT pathway, and the STAT pathway. Based on the mechanism of RTR in the TME, many proposed routes to overcome RTR, several specifically target the TME including targeting fibroblast activation protein, exosomes management, nanomedicine, and immunotherapy. Many challenges in RT resistance still need to be further explored with emerging investigative methods, such as artificial intelligence, genetic technologies, and bioengineering. CONCLUSIONS The complex interactions between RT and TME significantly affect the efficiency of RT. Novel approaches to overcome this clinical difficulty are promising, which needs future work to further explore and identify better treatment strategies.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinrui Li
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
| | - Siang Boon Koh
- Faculty of Health and Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Uzoamaka Okoli
- Division of Surgery & Interventional Science, University College London, London, UK; Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu, Nigeria
| | - Alisha Pati-Alam
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Eduardo Mota
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region of China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Division of Surgery & Interventional Science, University College London, London, UK.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, UK.
| |
Collapse
|
4
|
Wang Y, Yang B, Liu S, Song J, Zhang J, Chen X, Zheng N, He L, Cai W, Liu S. Semiconductor-mediated radiosensitizers: progress, challenges and perspectives. MATERIALS HORIZONS 2025. [PMID: 40035739 DOI: 10.1039/d4mh01703j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Radiotherapy has become one indispensable treatment strategy for treating malignant tumors. However, the therapeutic effect of radiotherapy is limited due to the low sensitivity and large side effects of existing radiosensitizers. The rapid development of nanotechnology has created opportunities for various novel kinds of radiosensitizers with excellent radiosensitivity to sprout recently. In particular, due to the ease of modification and potential utilization capacity for a multifunctional radiotherapy platform, semiconductor radiosensitizers have attracted more and more attention. Recently, many novel semiconductor based radiosensitizers have been reported, which provides new ideas for the improvement of radiotherapy efficacy. To make further breakthroughs in semiconductor radiosensitizers, a systematic review is urgently needed and is herein provided. This review first elaborates on the principle of semiconductor induced radiosensitization, and then focuses on strategies such as doping and constructing heterojunctions to enhance the radiosensitivity of semiconductors. Next, it introduces in detail the principle and progress of different types of semiconductor radiosensitizers. Finally, challenges and perspectives of semiconductor radiosensitizers are proposed and discussed, offering guidance for future commercial applications of semiconductor radiosensitizers.
Collapse
Affiliation(s)
- Yunsong Wang
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Bocan Yang
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Shujuan Liu
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Jiahe Song
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Jinghuai Zhang
- College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin 150001, P. R. China
| | - Xiangqun Chen
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Nannan Zheng
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Liangcan He
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Wei Cai
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Shaoqin Liu
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, P. R. China
| |
Collapse
|
5
|
Sano K, Mizukami T, Tanaka T, Nagayama T, Nakata M, Yamasaki M, Fukada T, Haratake Y, Munekane M, Yamasaki T, Mukai T. Intravenous administration of 90Y-labeled polyoxazoline combined with tumor heating potently inhibits tumor growth in mice. Int J Pharm 2025; 669:125103. [PMID: 39708850 DOI: 10.1016/j.ijpharm.2024.125103] [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: 08/24/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The basic requirements for the development of radiopharmaceuticals for radionuclide therapy of tumors include marked tumor-specific accumulation and long-term intratumoral retention. We have previously reported an indium-111 (111In)-labeled thermoresponsive polymer (polyoxazoline (POZ)) that is soluble at body temperature with rapid clearance from normal tissues but self-aggregates in the tumor upon tumor heating treatment. POZ accumulated in the tumor via self-aggregation under hyperthermic conditions and was retained after stopping heat exposure. In this study, we investigated the cellular uptake of 111In-labeled POZ and the antitumor effects of radionuclide therapy using yttrium-90 (90Y)-labeled POZ in combination with tumor heating. A POZ derivative with a lower critical solution temperature (LCST) of 38 °C was efficiently taken up by Colon-26 tumor cells at temperatures above the LCST via phagocytosis. In the therapeutic study using 90Y-labeled POZ, a marked dose-dependent therapeutic effect of radioactivity was observed in the groups treated with 90Y-POZ combined with tumor heating with no obvious systemic side effects. These results demonstrated that thermoresponsive 90Y-labeled POZ showed a potent therapeutic effect in combination with tumor heating, suggesting the usefulness for radionuclide therapy of tumors.
Collapse
Affiliation(s)
- Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan.
| | - Tatsuya Mizukami
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Toshie Tanaka
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Tomono Nagayama
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Momoka Nakata
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Mizuho Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Tomoya Fukada
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Yoshie Haratake
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| |
Collapse
|
6
|
Meher N, Bidkar AP, Wadhwa A, Bobba KN, Dhrona S, Dasari C, Mu C, Fong COY, Cámara JA, Ali U, Basak M, Bulkley D, Steri V, Fontaine SD, Zhu J, Oskowitz A, Aggarwal RR, Sriram R, Chou J, Wilson DM, Seo Y, Santi DV, Ashley GW, VanBrocklin HF, Flavell RR. PET Imaging Using 89Zr-Labeled StarPEG Nanocarriers Reveals Heterogeneous Enhanced Permeability and Retention in Prostate Cancer. Mol Cancer Ther 2025; 24:141-151. [PMID: 39331510 DOI: 10.1158/1535-7163.mct-24-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/05/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
The enhanced permeability and retention (EPR) effect controls passive nanodrug uptake in tumors and may provide a high tumor payload with prolonged retention for cancer treatment. However, EPR-mediated tumor uptake and distribution vary by cancer phenotype. Thus, we hypothesized that a companion PET imaging surrogate may benefit EPR-mediated therapeutic drug delivery. We developed two 89Zr-radiolabeled nanocarriers based on 4-armed starPEG40kDa with or without talazoparib (TLZ), a potent PARP inhibitor, as surrogates for the PEG-TLZ4 therapeutic scaffold. For PET imaging, PEG-DFB4 and PEG-DFB1-TLZ3 were radiolabeled with 89Zr by replacing one or all four copis of TLZ on PEG-TLZ4 with deferoxamine B (DFB). The radiolabeled nanodrugs [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 were tested in vivo in prostate cancer subcutaneous (s.c.) xenografts (22Rv1, LTL-545, and LTL-610) and 22Rv1 metastatic models. Their EPR-mediated tumoral uptake and penetration was compared with CT26, a known EPR-high cell line. MicroPET/CT images, organ biodistribution, and calculated kinetic parameters showed high uptake in CT26 and LTL-545 and moderate to low uptake in LTL-610 and 22Rv1. MicroPET/CT and high-resolution autoradiographic images showed nanocarrier penetration into highly permeable CT26, but heterogeneous peripheral accumulation was observed in LTL-545, LTL-610, and 22Rv1 s.c. xenografts and metastatic tumors. CD31 staining of tumor sections showed homogenous vascular development in CT26 tumors and heterogeneity in other xenografts. Both [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 showed similar accumulation and distribution in s.c. and metastatic tumor models. Both nanocarriers can measure tumor model passive uptake heterogeneity. Although heterogeneous, prostate cancer xenografts had low EPR. These starPEG nanocarriers could be used as PET imaging surrogates to predict drug delivery and efficacy.
Collapse
Affiliation(s)
- Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Chandrashekhar Dasari
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, California
| | - Changhua Mu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Cyril O Y Fong
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Juan A Cámara
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Umama Ali
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Megha Basak
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - David Bulkley
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | - Jun Zhu
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Adam Oskowitz
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, California
| | - Rahul R Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Jonathan Chou
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | | | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| |
Collapse
|
7
|
Suwannasing C, Suwannasom N, Soonthornchookiat P, Srisai P, Pattaweerakul C, Kothan S, Prapan A. The potential of HSA-stabilized zinc oxide nanoparticles as radiosensitizers to enhance the cytotoxic effects and radiosensitivity of cervical cancer cells. Cancer Nanotechnol 2024; 15:59. [DOI: 10.1186/s12645-024-00298-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/14/2024] [Indexed: 02/02/2025] Open
|
8
|
Sabu A, Kandel M, Sarma RR, Ramesan L, Roy E, Sharmila R, Chiu HC. Heterojunction semiconductor nanocatalysts as cancer theranostics. APL Bioeng 2024; 8:041502. [PMID: 39381587 PMCID: PMC11459490 DOI: 10.1063/5.0223718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
Cancer nanotechnology is a promising area of cross-disciplinary research aiming to develop facile, effective, and noninvasive strategies to improve cancer diagnosis and treatment. Catalytic therapy based on exogenous stimulus-responsive semiconductor nanomaterials has shown its potential to address the challenges under the most global medical needs. Semiconductor nanocatalytic therapy is usually triggered by the catalytic action of hot electrons and holes during local redox reactions within the tumor, which represent the response of nontoxic semiconductor nanocatalysts to pertinent internal or external stimuli. However, careful architecture design of semiconductor nanocatalysts has been the major focus since the catalytic efficiency is often limited by facile hot electron/hole recombination. Addressing these challenges is vital for the progress of cancer catalytic therapy. In recent years, diverse strategies have been developed, with heterojunctions emerging as a prominent and extensively explored method. The efficiency of charge separation under exogenous stimulation can be heightened by manipulating the semiconducting performance of materials through heterojunction structures, thereby enhancing catalytic capabilities. This review summarizes the recent applications of exogenous stimulus-responsive semiconducting nanoheterojunctions for cancer theranostics. The first part of the review outlines the construction of different heterojunction types. The next section summarizes recent designs, properties, and catalytic mechanisms of various semiconductor heterojunctions in tumor therapy. The review concludes by discussing the challenges and providing insights into their prospects within this dynamic and continuously evolving field of research.
Collapse
Affiliation(s)
- Arjun Sabu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Manoj Kandel
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ritwick Ranjan Sarma
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Lakshminarayan Ramesan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ekta Roy
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ramalingam Sharmila
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Cheng Chiu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
9
|
Inam A, Zhang S, Zhang S, Wu D. AQ4N nanocomposites for hypoxia-associated tumor combination therapy. Biomater Sci 2024; 12:5883-5911. [PMID: 39431892 DOI: 10.1039/d4bm00883a] [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: 10/22/2024]
Abstract
Hypoxia in solid tumors increases their invasiveness and resistance to therapy, presenting a formidable obstacle in tumor therapy. The hypoxia prodrug banoxantrone (AQ4N) undergoes conversion into its topoisomerase II inhibitor form AQ4 under hypoxic conditions, which inhibits tumor cells while leaving normal cells unharmed. Numerous studies have found that AQ4N significantly enhances the tumor effect while minimizing toxicity to normal tissues when combined with other drugs or therapeutic approaches. Thus, to maximize AQ4N's effectiveness, co-delivery of AQ4N with other therapeutic agents to the tumor site is paramount, leading to the development of multifunctional multicomponent AQ4N nanocomposites thereby emerging as promising candidates for combination therapy in tumor treatment. However, currently there is a lack of systematic analysis and reviews focusing on AQ4N. Herein, this review provides a comprehensive retrospect and analysis of the recent advancements in AQ4N nanocomposites. Specifically, we discuss the synergistic effects observed when AQ4N is combined with chemotherapeutic drugs, radiotherapy, phototherapy, starvation, sonodynamic therapy and immunotherapy in preclinical models. Moreover, the advantages, limitations, and future perspectives of different AQ4N nanocomposites are highlighted, providing researchers from diverse fields with novel insights into tumor treatment.
Collapse
Affiliation(s)
- Amrah Inam
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Shuo Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Shuai Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| |
Collapse
|
10
|
Pires ICB, Shuchi SI, Tostes BDVA, Santos DKDDN, Burnett WL, Leonce BC, Harvey OR, Coffer JL, de Sousa Filho IA, de Athayde-Filho PF, Junior SA, Mathis JM. Theranostics Using MCM-41-Based Mesoporous Silica Nanoparticles: Integrating Magnetic Resonance Imaging and Novel Chemotherapy for Breast Cancer Treatment. Int J Mol Sci 2024; 25:8097. [PMID: 39125669 PMCID: PMC11311303 DOI: 10.3390/ijms25158097] [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: 06/21/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Advanced breast cancer remains a significant oncological challenge, requiring new approaches to improve clinical outcomes. This study investigated an innovative theranostic agent using the MCM-41-NH2-DTPA-Gd3⁺-MIH nanomaterial, which combined MRI imaging for detection and a novel chemotherapy agent (MIH 2.4Bl) for treatment. The nanomaterial was based on the mesoporous silica type, MCM-41, and was optimized for drug delivery via functionalization with amine groups and conjugation with DTPA and complexation with Gd3+. MRI sensitivity was enhanced by using gadolinium-based contrast agents, which are crucial in identifying early neoplastic lesions. MIH 2.4Bl, with its unique mesoionic structure, allows effective interactions with biomolecules that facilitate its intracellular antitumoral activity. Physicochemical characterization confirmed the nanomaterial synthesis and effective drug incorporation, with 15% of MIH 2.4Bl being adsorbed. Drug release assays indicated that approximately 50% was released within 8 h. MRI phantom studies demonstrated the superior imaging capability of the nanomaterial, with a relaxivity significantly higher than that of the commercial agent Magnevist. In vitro cellular cytotoxicity assays, the effectiveness of the nanomaterial in killing MDA-MB-231 breast cancer cells was demonstrated at an EC50 concentration of 12.6 mg/mL compared to an EC50 concentration of 68.9 mg/mL in normal human mammary epithelial cells (HMECs). In vivo, MRI evaluation in a 4T1 syngeneic mouse model confirmed its efficacy as a contrast agent. This study highlighted the theranostic capabilities of MCM-41-NH2-DTPA-Gd3⁺-MIH and its potential to enhance breast cancer management.
Collapse
Affiliation(s)
- Indira C. B. Pires
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - Samia I. Shuchi
- School of Biomedical Sciences, Departments of Microbiology, Immunology, and Genetics and Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Braulio de V. A. Tostes
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - Dayane K. D. do N. Santos
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - William L. Burnett
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Burke C. Leonce
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Omar R. Harvey
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Jeffery L. Coffer
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, TX 76109, USA (B.C.L.); (O.R.H.); (J.L.C.)
| | - Idio Alves de Sousa Filho
- Institute of Chemistry, Federal Rural University of Rio de Janeiro, Rio de Janeiro 23890-000, RJ, Brazil;
| | | | - Severino A. Junior
- Department of Chemistry, Federal University of Pernambuco, Recife 50670-901, PE, Brazil; (I.C.B.P.); (B.d.V.A.T.); (D.K.D.d.N.S.)
| | - J. Michael Mathis
- School of Biomedical Sciences, Departments of Microbiology, Immunology, and Genetics and Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| |
Collapse
|
11
|
Yang G, Cao Y, Yang X, Cui T, Tan NZV, Lim YK, Fu Y, Cao X, Bhandari A, Enikeev M, Efetov S, Balaban V, He M. Advancements in nanomedicine: Precision delivery strategies for male pelvic malignancies - Spotlight on prostate and colorectal cancer. Exp Mol Pathol 2024; 137:104904. [PMID: 38788248 DOI: 10.1016/j.yexmp.2024.104904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Pelvic malignancies consistently pose significant global health challenges, adversely affecting the well-being of the male population. It is anticipated that clinicians will continue to confront these cancers in their practice. Nanomedicine offers promising strategies that revolutionize the treatment of male pelvic malignancies by providing precise delivery methods that aim to improve the efficacy of therapeutic outcomes while minimizing side effects. Nanoparticles are designed to encapsulate therapeutic agents and selectively target cancer cells. They can also be loaded with theragnostic agents, enabling multifunctional capabilities. OBJECTIVE This review aims to summarize the latest nanomedicine research into clinical applications, focusing on nanotechnology-based treatment strategies for male pelvic malignancies, encompassing chemotherapy, radiotherapy, immunotherapy, and other cutting-edge therapies. The review is structured to assist physicians, particularly those with limited knowledge of biochemistry and bioengineering, in comprehending the functionalities and applications of nanomaterials. METHODS Multiple databases, including PubMed, the National Library of Medicine, and Embase, were utilized to locate and review recently published articles on advancements in nano-drug delivery for prostate and colorectal cancers. CONCLUSION Nanomedicine possesses considerable potential in improving therapeutic outcomes and reducing adverse effects for male pelvic malignancies. Through precision delivery methods, this emerging field presents innovative treatment modalities to address these challenging diseases. Nevertheless, the majority of current studies are in the preclinical phase, with a lack of sufficient evidence to fully understand the precise mechanisms of action, absence of comprehensive pharmacotoxicity profiles, and uncertainty surrounding long-term consequences.
Collapse
Affiliation(s)
- Guodong Yang
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Te Cui
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Yuen Kai Lim
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yu Fu
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xinren Cao
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aanchal Bhandari
- HBT Medical College and Dr. R N Cooper Municipal General Hospital, Mumbai, India
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Sergey Efetov
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir Balaban
- Clinic of Coloproctology and Minimally Invasive Surgery, Sechenov University, Moscow, Russia
| | - Mingze He
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
| |
Collapse
|
12
|
Chen Z, Liao Z, Liu M, Lin F, Chen S, Wang G, Zheng Z, Liu B, Li C, Wang Z, Chen T, Huang H, Liao Q, Cui W. Nucleus Pulposus-Targeting Nanocarriers Facilitate Mirna-Based Therapeutics for Intervertebral Disc Degeneration. Adv Healthc Mater 2023; 12:e2301337. [PMID: 37625164 DOI: 10.1002/adhm.202301337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/23/2023] [Indexed: 08/27/2023]
Abstract
Intervertebral disc degeneration (IDD) is a common cause of low back pain. Understanding its molecular mechanisms is the basis for developing specific treatment. To demonstrate that miR-22-3p is critical in the regulation of IDD, miRNA microarray analyses are conducted in conjunction with in vivo and in vitro experiments. The miR-22-3p knockout (KO) mice show a marked decrease in the histological scores. Bioinformatic analysis reveals that miR-22-3p plays a mechanistic role in the development of IDD by targeting SIRT1, which in turn activates the JAK1/STAT3 signaling pathway. This is confirmed by a luciferase reporter assay and western blot analysis. Therapeutically, the delivery of miR-22-3p inhibitors and mimics through the synthesized nanoparticles in the IDD model alleviates and aggravates IDD, respectively. The nanocarriers enhance transportation of miR-22-3p to nucleus pulposus cells, thus enabling the in vivo inhibition of miR-22-3p for therapeutic purposes and consequently promoting the development of miRNA-specific drugs for IDD.
Collapse
Affiliation(s)
- Zhonghui Chen
- Orthopaedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350000, China
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
- Orthopaedic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430000, China
| | - Zhong Liao
- Orthopaedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350000, China
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Ming Liu
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, 350000, China
| | - Fengfei Lin
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Shunyou Chen
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Geng Wang
- Department of Pharmacology, School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, Fujian, 350000, China
| | - Zhong Zheng
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Boling Liu
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Chaoxiong Li
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Zheqiang Wang
- Department of Sport's Medicine, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, 350000, China
| | - Tianlai Chen
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Hongzhe Huang
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| | - Qi Liao
- Orthopaedic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430000, China
| | - Weiliang Cui
- Orthopaedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350000, China
- Orthopaedic Surgery, Fuzhou Second Hospital, Fuzhou, Fujian, 350000, China
| |
Collapse
|
13
|
Babu B, Stoltz SA, Mittal A, Pawar S, Kolanthai E, Coathup M, Seal S. Inorganic Nanoparticles as Radiosensitizers for Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2873. [PMID: 37947718 PMCID: PMC10647410 DOI: 10.3390/nano13212873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Nanotechnology has expanded what can be achieved in our approach to cancer treatment. The ability to produce and engineer functional nanoparticle formulations to elicit higher incidences of tumor cell radiolysis has resulted in substantial improvements in cancer cell eradication while also permitting multi-modal biomedical functionalities. These radiosensitive nanomaterials utilize material characteristics, such as radio-blocking/absorbing high-Z atomic number elements, to mediate localized effects from therapeutic irradiation. These materials thereby allow subsequent scattered or emitted radiation to produce direct (e.g., damage to genetic materials) or indirect (e.g., protein oxidation, reactive oxygen species formation) damage to tumor cells. Using nanomaterials that activate under certain physiologic conditions, such as the tumor microenvironment, can selectively target tumor cells. These characteristics, combined with biological interactions that can target the tumor environment, allow for localized radio-sensitization while mitigating damage to healthy cells. This review explores the various nanomaterial formulations utilized in cancer radiosensitivity research. Emphasis on inorganic nanomaterials showcases the specific material characteristics that enable higher incidences of radiation while ensuring localized cancer targeting based on tumor microenvironment activation. The aim of this review is to guide future research in cancer radiosensitization using nanomaterial formulations and to detail common approaches to its treatment, as well as their relations to commonly implemented radiotherapy techniques.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Samantha Archer Stoltz
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Melanie Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA;
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
14
|
Grissi C, Taverna Porro M, Perona M, Atia M, Negrin L, Moreno MS, Sacanell J, Olivera MS, Del Grosso M, Durán H, Ibañez IL. Superparamagnetic iron oxide nanoparticles induce persistent large foci of DNA damage in human melanoma cells post-irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2023:10.1007/s00411-023-01037-0. [PMID: 37452828 DOI: 10.1007/s00411-023-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 06/25/2023] [Indexed: 07/18/2023]
Abstract
The synergy of superparamagnetic iron oxide nanoparticles (SPIONs) and ionizing radiation (IR), attributed to reactive oxygen species (ROS) and DNA double-strand breaks (DSBs) increase, was widely investigated in different cancers, but scarcely in melanoma. Herein, SPIONs were evaluated as radiosensitizers in A-375 human melanoma cells. Moreover, the effect of the combined treatment of SPIONs and gamma irradiation (SPIONs-IR) was assessed at the DNA level, where DSBs induction and their repair capacity were studied. SPIONs were synthesized, stabilized by poly(ethylene glycol) methyl ether and physicochemically characterized by high resolution-transmission electron microscopy (HR-TEM), X-ray diffraction and magnetometry and dynamic light scattering. The obtained nanoparticles showing superparamagnetic behavior and low dispersion in shape and sizes were tested in A-375 cells. The intracellular internalization of SPIONs was verified by HR-TEM and quantified by inductively coupled plasma atomic emission spectroscopy. Cells treated with SPIONs exhibited high ROS levels without associated cytotoxicity. Next, a significant radiosensitization in SPIONs-IR vs. control (IR) cells was demonstrated at 1 Gy of gamma radiation. Furthermore, a decreased DSBs repair capacity in SPIONs-IR vs. IR-treated cells was evidenced by the size increase of persistent phosphorylated H2AX foci at 24 h post-irradiation. In conclusion, these nanoparticles show the potential to radiosensitize melanoma cells by the induction of unrepairable DNA damage.
Collapse
Affiliation(s)
- Cecilia Grissi
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Marisa Taverna Porro
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos Aires, Junín 954 (C1113AAD), Ciudad Autónoma de Buenos Aires, Argentina
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Campus Miguelete (B1650KNA), San Martín, Provincia de Buenos Aires, Argentina
| | - Marina Perona
- División Bioquímica Nuclear, Departamento de Radiobiología, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Mariel Atia
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Lara Negrin
- Laboratorio de Radiobiología y Biodosimetría, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Centro de Medicina Nuclear y Radioterapia - Instituto de Tecnologías Nucleares Para La Salud (INTECNUS), Av. Bustillo Km. 9,5 (R8402AGP), S.C. de Bariloche, Río Negro, Argentina
| | - M Sergio Moreno
- Instituto de Nanociencia y Nanotecnología (INN), Comisión Nacional de Energía Atómica (CNEA) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Bariloche, Centro Atómico Bariloche, Av. Bustillo Km. 9,5 (R8402AGP), S.C. de Bariloche, Río Negro, Argentina
| | - Joaquín Sacanell
- Departamento de Física de la Materia Condensada, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - María Silvina Olivera
- Departamento Coordinación BNCT, Comisión Nacional de Energía Atómica (CNEA), Centro Atómico Constituyentes, Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Mariela Del Grosso
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Hebe Durán
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina.
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Campus Miguelete (B1650KNA), San Martín, Provincia de Buenos Aires, Argentina.
| | - Irene L Ibañez
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina.
| |
Collapse
|
15
|
Arif M, Nawaz AF, Ullah khan S, Mueen H, Rashid F, Hemeg HA, Rauf A. Nanotechnology-based radiation therapy to cure cancer and the challenges in its clinical applications. Heliyon 2023; 9:e17252. [PMID: 37389057 PMCID: PMC10300336 DOI: 10.1016/j.heliyon.2023.e17252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023] Open
Abstract
Radiation therapy against cancer frequently fails to attain the desired outcomes because of several restricting aspects. Radiation therapy is not a targeted antitumor treatment, and it poses serious threats to normal tissues as well. In many cases, some inherent features of tumors make them resistant to radiation therapy. Several nanoparticles have shown the capacity to upgrade the viability of radiation treatment because they can directly interact with ionizing radiation to increase cellular radiation sensitivity. Several types of nanomaterials have been investigated as radio-sensitizers, to improve the efficacy of radiotherapy and overcome radio-resistance including, metal-based nanoparticles, quantum dots, silica-based nanoparticles, polymeric nanoparticles, etc. Despite all this research and development, certain challenges associated with the exploitation of nanoparticles to enhance and improve radiation therapy for cancer treatment are encountered. Potential applications of nanoparticles as radiosensitizers is hindered by the difficulties associated with ensuring their production at a large scale with improved characterizations and because of certain biological challenges. By overcoming the shortcomings of nanoparticles like working on the pharmacokinetics, and physical and chemical characterization, the therapy can be improved. It is expected that in the future more knowledge will be available regarding nanoparticles and their clinical efficacy, leading to the successful development of nanotechnology-based radiation therapies for a variety of cancers. This review highlights the limitations of conventional radiotherapy in cancer treatment and explores the potential of nanotechnology, specifically the use of nanomaterials, to overcome these challenges. It discusses the concept of using nanomaterials to enhance the effectiveness of radiation therapy and provides an overview of different types of nanomaterials and their beneficial properties. The review emphasizes the need to address the obstacles and limitations associated with the application of nanotechnology in cancer radiation therapy for successful clinical translation.
Collapse
Affiliation(s)
- Muhammad Arif
- Department of Plant Biology and Ecology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Ayesha Fazal Nawaz
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Centre (NARC), Islamabad, Pakistan
| | - Shahid Ullah khan
- Department of Biochemistry, Women Medical and Dental College, Khyber Medical University KPK, Pakistan
| | - Hasnat Mueen
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Fizza Rashid
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Al-Medinah Al-Monawara Postcode, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
16
|
Haque M, Shakil MS, Mahmud KM. The Promise of Nanoparticles-Based Radiotherapy in Cancer Treatment. Cancers (Basel) 2023; 15:cancers15061892. [PMID: 36980778 PMCID: PMC10047050 DOI: 10.3390/cancers15061892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Radiation has been utilized for a long time for the treatment of cancer patients. However, radiotherapy (RT) has many constraints, among which non-selectivity is the primary one. The implementation of nanoparticles (NPs) with RT not only localizes radiation in targeted tissue but also provides significant tumoricidal effect(s) compared to radiation alone. NPs can be functionalized with both biomolecules and therapeutic agents, and their combination significantly reduces the side effects of RT. NP-based RT destroys cancer cells through multiple mechanisms, including ROS generation, which in turn damages DNA and other cellular organelles, inhibiting of the DNA double-strand damage-repair system, obstructing of the cell cycle, regulating of the tumor microenvironment, and killing of cancer stem cells. Furthermore, such combined treatments overcome radioresistance and drug resistance to chemotherapy. Additionally, NP-based RT in combined treatments have shown synergistic therapeutic benefit(s) and enhanced the therapeutic window. Furthermore, a combination of phototherapy, i.e., photodynamic therapy and photothermal therapy with NP-based RT, not only reduces phototoxicity but also offers excellent therapeutic benefits. Moreover, using NPs with RT has shown promise in cancer treatment and shown excellent therapeutic outcomes in clinical trials. Therefore, extensive research in this field will pave the way toward improved RT in cancer treatment.
Collapse
Affiliation(s)
- Munima Haque
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Md Salman Shakil
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Kazi Mustafa Mahmud
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| |
Collapse
|
17
|
Han G, Xiang S, Jiang K, Zhang W, Weng Q. Design of size uniform and controllable covalent organic framework nanoparticles for high-performance anticancer drug delivery. J Biomater Appl 2023; 37:1376-1383. [PMID: 36472170 DOI: 10.1177/08853282221144526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Covalent organic frameworks (COFs) receive much attention in biomedicine because of their unique adsorption, optical and biological properties, as well as highly variable structures. However, preparation of nanosized COFs with uniform and controllable size is still a challenge. Herein, we develop a facile interfacial method to prepare the COF nanoparticles (COFNPs) with the uniform size of 30-50 nm from p-benzoquinone (BQ) and 4-[1,2,2-tris(4-aminophenyl)ethenyl]aniline (TPEA) by Michael addition. The TPEA-BQ COFNPs show positive zeta potential and effectively load the hydrophobic anticancer drug camptothecin (CPT) with the capacity of up to 127wt%, and remarkably improved the CPT dispersibility in water due to the retention of quinone structure. In vitro assay reveals CPT@ TPEA-BQ significantly reduced cell viability to 29% after 24 h incubation, much lower than that of free CPT (51%) at the same concentration of 10 μg mL-1. Further in vivo experiment confirms the high anticancer drug delivery performance of the designed TPEA-BQ COFNPs. After 20 days of injection treatment, the CPT loaded in TPEA-BQ COFNPs inhibits the tumor growth by 60%, much higher than that of free CPT group (23%). This work demonstrates the feasibility to design advanced drug delivery systems based on highly structure-tunable COF system.
Collapse
Affiliation(s)
- Guangze Han
- College of Materials Science and Engineering, 12569Hunan University, Changsha, China
| | - Shuo Xiang
- College of Materials Science and Engineering, 12569Hunan University, Changsha, China
| | - Kang Jiang
- College of Materials Science and Engineering, 12569Hunan University, Changsha, China
| | - Wei Zhang
- College of Materials Science and Engineering, 12569Hunan University, Changsha, China
| | - Qunhong Weng
- College of Materials Science and Engineering, 12569Hunan University, Changsha, China
| |
Collapse
|
18
|
Nanoparticles in Clinical Trials: Analysis of Clinical Trials, FDA Approvals and Use for COVID-19 Vaccines. Int J Mol Sci 2023; 24:ijms24010787. [PMID: 36614230 PMCID: PMC9821409 DOI: 10.3390/ijms24010787] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
Nanoparticles are heterologous small composites that are usually between 1 and 100 nanometers in size. They are applied in many areas of medicine with one of them being drug delivery. Nanoparticles have a number of advantages as drug carriers which include reduced toxic effects, increased bioavailability, and their ability to be modified for specific tissues or cells. Due to the exciting development of nanotechnology concomitant with advances in biotechnology and medicine, the number of clinical trials devoted to nanoparticles for drug delivery is growing rapidly. Some nanoparticles, lipid-based types, in particular, played a crucial role in the developing and manufacturing of the two COVID-19 vaccines-Pfizer and Moderna-that are now being widely used. In this analysis, we provide a quantitative survey of clinical trials using nanoparticles during the period from 2002 to 2021 as well as the recent FDA-approved drugs (since 2016). A total of 486 clinical trials were identified using the clinicaltrials.gov database. The prevailing types of nanoparticles were liposomes (44%) and protein-based formulations (26%) during this period. The most commonly investigated content of the nanoparticles were paclitaxel (23%), metals (11%), doxorubicin (9%), bupivacaine and various vaccines (both were 8%). Among the FDA-approved nanoparticle drugs, polymeric (29%), liposomal (22%) and lipid-based (21%) drugs were the most common. In this analysis, we also discuss the differential development of the diverse groups of nanoparticles and their content, as well as the underlying factors behind the trends.
Collapse
|
19
|
Yang F, He Q, Dai X, Zhang X, Song D. The potential role of nanomedicine in the treatment of breast cancer to overcome the obstacles of current therapies. Front Pharmacol 2023; 14:1143102. [PMID: 36909177 PMCID: PMC9992554 DOI: 10.3389/fphar.2023.1143102] [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: 01/12/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignant tumor among women in the world. BC is the heterogeneous tumor with different subtypes including luminal A-like, luminal B-like (HER2-/HER2+), HER2 enriched, and triple-negative BC. The therapeutic strategies including surgery, chemotherapy, radiotherapy, targeted therapy, and endocrine therapy are well developed and commonly used in the treatment of BC. However, some adverse effects of these conventional treatments limited their wide application in clinical. Therefore, it is necessary to develop more safe and more efficient individualized treatment strategies of the BC. Nanomedicine, as the most promising strategy for controlled and targeted drug delivery, is widely used in multiple aspects of cancer therapy. Importantly, accumulative evidences show that nanomedicine has achieved good outcomes in the treatment of BC and a huge amount of BC patients benefited from the nanomedicine related treatments. In this review, we summarized and discussed the major problems occurred during the administration of conventional treatment strategies for BC and the potential roles of nanomedicine in promoting the treatment efficacy of BC by overcoming obstacles of current treatment of BC.
Collapse
Affiliation(s)
- Fan Yang
- Breast Surgery Department of General Surgery, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Qingjie He
- Breast Surgery Department of General Surgery, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Dong Song
- Breast Surgery Department of General Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Sun S, Zhao Y, Wang J, Pei R. Lanthanide-based MOFs: synthesis approaches and applications in cancer diagnosis and therapy. J Mater Chem B 2022; 10:9535-9564. [PMID: 36385652 DOI: 10.1039/d2tb01884e] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metal-organic frameworks (MOFs) have attracted considerable attention as emerging nanomaterials. Based on their tunable size, high porosity, and large specific surface area, MOFs have a wide range of applications in the fields of chemistry, energy, and biomedicine. However, the MOF materials obtained from lanthanides with a unique electronic configuration as inorganic building units have unique properties such as optics, magnetism, and radioactivity. In this study, various synthetic methods for preparing MOF materials using lanthanides as inorganic building units are described. Combined with the characteristics of lanthanides, their application prospects of lanthanide-based MOFs in tumor diagnosis and treatment are emphasized. The authors hope to provide methodological reference for the construction of MOF materials of rare-earth elements, and to provide ideas and inspiration for their practical applications in the field of biomedicine.
Collapse
Affiliation(s)
- Shengkai Sun
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Jine Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China. .,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China. .,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
21
|
Meher N, Ashley GW, Bidkar AP, Dhrona S, Fong C, Fontaine SD, Beckford Vera DR, Wilson DM, Seo Y, Santi DV, VanBrocklin HF, Flavell RR. Prostate-Specific Membrane Antigen Targeted Deep Tumor Penetration of Polymer Nanocarriers. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50569-50582. [PMID: 36318757 PMCID: PMC9673064 DOI: 10.1021/acsami.2c15095] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/24/2022] [Indexed: 05/05/2023]
Abstract
Tumoral uptake of large-size nanoparticles is mediated by the enhanced permeability and retention (EPR) effect, with variable accumulation and heterogenous tumor tissue penetration depending on the tumor phenotype. The performance of nanocarriers via specific targeting has the potential to improve imaging contrast and therapeutic efficacy in vivo with increased deep tissue penetration. To address this hypothesis, we designed and synthesized prostate cancer-targeting starPEG nanocarriers (40 kDa, 15 nm), [89Zr]PEG-(DFB)3(ACUPA)1 and [89Zr]PEG-(DFB)1(ACUPA)3, with one or three prostate-specific membrane antigen (PSMA)-targeting ACUPA ligands. The in vitro PSMA binding affinity and in vivo pharmacokinetics of the targeted nanocarriers were compared with a nontargeted starPEG, [89Zr]PEG-(DFB)4, in PSMA+ PC3-Pip and PSMA- PC3-Flu cells, and xenografts. Increasing the number of ACUPA ligands improved the in vitro binding affinity of PEG-derived polymers to PC3-Pip cells. While both PSMA-targeted nanocarriers significantly improved tissue penetration in PC3-Pip tumors, the multivalent [89Zr]PEG-(DFB)1(ACUPA)3 showed a remarkably higher PC3-Pip/blood ratio and background clearance. In contrast, the nontargeted [89Zr]PEG-(DFB)4 showed low EPR-mediated accumulation with poor tumor tissue penetration. Overall, ACUPA conjugated targeted starPEGs significantly improve tumor retention with deep tumor tissue penetration in low EPR PC3-Pip xenografts. These data suggest that PSMA targeting with multivalent ACUPA ligands may be a generally applicable strategy to increase nanocarrier delivery to prostate cancer. These targeted multivalent nanocarriers with high tumor binding and low healthy tissue retention could be employed in imaging and therapeutic applications.
Collapse
Affiliation(s)
- Niranjan Meher
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - Gary W. Ashley
- ProLynx
Inc., San Francisco, California 94158, United States
| | - Anil P. Bidkar
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - Suchi Dhrona
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - Cyril Fong
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | | | - Denis R. Beckford Vera
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - David M. Wilson
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Youngho Seo
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Daniel V. Santi
- ProLynx
Inc., San Francisco, California 94158, United States
| | - Henry F. VanBrocklin
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Robert R. Flavell
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, California 94158-2517, United States
| |
Collapse
|
22
|
Ayana G, Ryu J, Choe SW. Ultrasound-Responsive Nanocarriers for Breast Cancer Chemotherapy. MICROMACHINES 2022; 13:1508. [PMID: 36144131 PMCID: PMC9503784 DOI: 10.3390/mi13091508] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 05/13/2023]
Abstract
Breast cancer is the most common type of cancer and it is treated with surgical intervention, radiotherapy, chemotherapy, or a combination of these regimens. Despite chemotherapy's ample use, it has limitations such as bioavailability, adverse side effects, high-dose requirements, low therapeutic indices, multiple drug resistance development, and non-specific targeting. Drug delivery vehicles or carriers, of which nanocarriers are prominent, have been introduced to overcome chemotherapy limitations. Nanocarriers have been preferentially used in breast cancer chemotherapy because of their role in protecting therapeutic agents from degradation, enabling efficient drug concentration in target cells or tissues, overcoming drug resistance, and their relatively small size. However, nanocarriers are affected by physiological barriers, bioavailability of transported drugs, and other factors. To resolve these issues, the use of external stimuli has been introduced, such as ultrasound, infrared light, thermal stimulation, microwaves, and X-rays. Recently, ultrasound-responsive nanocarriers have become popular because they are cost-effective, non-invasive, specific, tissue-penetrating, and deliver high drug concentrations to their target. In this paper, we review recent developments in ultrasound-guided nanocarriers for breast cancer chemotherapy, discuss the relevant challenges, and provide insights into future directions.
Collapse
Affiliation(s)
- Gelan Ayana
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| | - Jaemyung Ryu
- Department of Optical Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| | - Se-woon Choe
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| |
Collapse
|
23
|
Wang Y, Zhang H, Liu Y, Younis MH, Cai W, Bu W. Catalytic radiosensitization: Insights from materials physicochemistry. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2022; 57:262-278. [PMID: 36425004 PMCID: PMC9681018 DOI: 10.1016/j.mattod.2022.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Radiotherapy is indispensable in clinical cancer treatment, but because both tumor and normal tissues have similar sensitivity to X-rays, their clinical curative effect is intrinsically limited. Advanced nanomaterials and nanotechnologies have been developed for radiotherapy sensitization, typically employing high atomic number (high-Z) materials to enhance the energy deposition of X-rays in tumor tissues, but the efficiency is largely limited by the toxicity of heavy metals. A new and promising approach for radiosensitization is catalytic radiosensitization, which takes advantage of the catalytic activity of nanomaterials triggered by radiation. The efficiency of catalytic radiosensitization can be greatly enhanced by electron modulation and energy conversion of nanocatalysts upon X-ray irradiation, further enhancing the clinical curative effect. In this review, we highlight the challenges and opportunities in cancer radiosensitization, discuss novel approaches to catalytic radiosensitization, and finally describe the development of catalytic radiosensitization based on an in-depth understanding of radio-nano interactions and catalysis-biological interactions.
Collapse
Affiliation(s)
- Ya Wang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
| | - Huilin Zhang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
| | - Muhsin H. Younis
- Department of Radiology and Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Weibo Cai
- Department of Radiology and Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| |
Collapse
|
24
|
Păduraru DN, Ion D, Niculescu AG, Mușat F, Andronic O, Grumezescu AM, Bolocan A. Recent Developments in Metallic Nanomaterials for Cancer Therapy, Diagnosing and Imaging Applications. Pharmaceutics 2022; 14:435. [PMID: 35214167 PMCID: PMC8874382 DOI: 10.3390/pharmaceutics14020435] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer continues to represent a global health concern, imposing an ongoing need to research for better treatment alternatives. In this context, nanomedicine seems to be the solution to existing problems, bringing unprecedented results in various biomedical applications, including cancer therapy, diagnosing, and imaging. As numerous studies have uncovered the advantageous properties of various nanoscale metals, this review aims to present metal-based nanoparticles that are most frequently employed for cancer applications. This paper follows the description of relevant nanoparticles made of metals, metal derivatives, hybrids, and alloys, further discussing in more detail their potential applications in cancer management, ranging from the delivery of chemotherapeutics, vaccines, and genes to ablative hyperthermia therapies and theranostic platforms.
Collapse
Affiliation(s)
- Dan Nicolae Păduraru
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (D.N.P.); (D.I.); (F.M.); (O.A.); (A.B.)
- Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
| | - Daniel Ion
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (D.N.P.); (D.I.); (F.M.); (O.A.); (A.B.)
- Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
| | - Florentina Mușat
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (D.N.P.); (D.I.); (F.M.); (O.A.); (A.B.)
- Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
| | - Octavian Andronic
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (D.N.P.); (D.I.); (F.M.); (O.A.); (A.B.)
- Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov No. 3, 50044 Bucharest, Romania
| | - Alexandra Bolocan
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (D.N.P.); (D.I.); (F.M.); (O.A.); (A.B.)
- Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
| |
Collapse
|
25
|
Omar AS. Nanoformulation Safety versus Toxicity; What do the Recent Studies Tell Us? INTERNATIONAL JOURNAL OF PHARMACEUTICAL RESEARCH AND ALLIED SCIENCES 2022. [DOI: 10.51847/spfpldpsvl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
26
|
Yilmaz H, Karakoc Y, Tumkaya L, Mercantepe T, Sevinc H, Yilmaz A, Yılmaz Rakıcı S. The protective effects of red ginseng and amifostine against renal damage caused by ionizing radiation. Hum Exp Toxicol 2022; 41:9603271221143029. [DOI: 10.1177/09603271221143029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
This study aimed to elucidate the effects of amifostine (ethyol) (AM), a synthetic radioprotector, and red ginseng (RG), a natural radioprotective agent, against the toxic effect of ionizing radiation (IR) on kidney tissues through changes in biochemical and histopathological parameters in addition to contributions to the use of amifostine and RG in clinical studies . Five groups were established: Group I (control, receiving only saline by gavage), Group II (IR only), and Group III (IR+AM, 200 mg/kg intraperitoneally (i.p.). Group IV (IR + RG, 200 mg/kg orally once a day for 4 weeks), and Group V (IR+RG+AM, 200 mg/kg orally once/day for 4 weeks before IR and 200 mg/kg AM administered (i.p.) 30 min before IR). All groups, except for the control group, were subject to 6-Gy whole-body IR in a single fraction. 24 h after irradiation, all animals were sacrificed under anesthesia. IR enhanced MDA, 8-OHdG, and caspase-3 expression while decreasing renal tissue GSH levels ( p < .05). Significant numbers of necrotic tubules together with diffuse vacuolization in proximal and distal tubule epithelial cells were also observed. The examination also revealed substantial brush boundary loss in proximal tubules as well as relatively unusual glomerular structures. While GSH levels significantly increased in the AM, RG, and AM+RG groups, a decrease in KHDS, MDA, 8-OHdG, and caspase-3 expression was observed, compared to the group subject to IR only ( p < .05). Therefore, reactive oxygen species-scavenging antioxidants may represent a promising treatment for avoiding kidney damage in patients receiving radiation.
Collapse
Affiliation(s)
- Hamit Yilmaz
- Department of Biophysics, Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Yunus Karakoc
- Department of Biophysics, Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University Rize, Turkey
| | - Hacer Sevinc
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University Rize, Turkey
| | - Adnan Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University Rize, Turkey
| | - Sema Yılmaz Rakıcı
- Department of Radiation Oncology, Faculty of Medicine, Recep Tayyip Erdogan University Rize, Turkey
| |
Collapse
|