1
|
Yılmaz E, Kacaroglu D, Ozden AK, Aydogan N. Gold nanoparticles decorated FOLFIRINOX loaded liposomes for synergistic therapy of pancreatic cancer. Int J Pharm 2025; 669:125067. [PMID: 39672312 DOI: 10.1016/j.ijpharm.2024.125067] [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: 08/21/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Pancreatic cancer is predicted to be the second highest cause of cancer deaths by 2030, with a mortality rate of 98 % and a 5-year survival rate of only 4-8 %. FOLFIRINOX which consists of four main ingredients has shown superior efficacy in treating patients with pancreatic cancer compared to other agents and combinations. However, toxicities have prevented full-dose use of FOLFIRINOX. In this study, we present the design of a liposome nanosystem that enables the sequential release of a drug combination that is called FOLFIRINOX using lipid-based nanosystem synergistic chemo/photothermal therapy approaches. The co-eccentric liposome allowed us to locate the drug molecules in different locations giving us the flexibility to release them in a selected order. Core liposome (L2) has a melting temperature of 53.63 °C, it was decorated by gold nanoparticle (L2@AuNP) to bring photothermal responsiveness. The outer liposome structure had a lower melting temperature, which facilitated the sequential release process. The efficacy of photothermal therapy for nanosystem was calculated. The results indicate that coating L2@AuNP nanostructure with L1 liposomes improves efficacy by stabilizing gold nanoparticles. FOLFIRINOX components are encapsulated in a concentric liposome structure according to the order of administration into the body. The concentric liposome structure enables the sequential release of multiple drugs due to the varying phase transition temperatures of the liposomes. The cytotoxic effect of these formulations was evaluated on Panc-1 pancreatic cancer cells; the lowest cell viability was obtained in 4 Liposome(L) under 5 min NIR irradiation. Combination therapy has a higher therapeutic efficacy (70.45 %) when compared to chemotherapy and photothermal therapy used separately. The study's results show the potential of combination therapies to improve therapeutic outcomes, providing a promising path for future research and clinical application.
Collapse
Affiliation(s)
- Emine Yılmaz
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey
| | - Demet Kacaroglu
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Ayse Kevser Ozden
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Nihal Aydogan
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey; Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara, 06800, Turkey.
| |
Collapse
|
2
|
Keum H, Cevik E, Kim J, Demirlenk YM, Atar D, Saini G, Sheth RA, Deipolyi AR, Oklu R. Tissue Ablation: Applications and Perspectives. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310856. [PMID: 38771628 PMCID: PMC11309902 DOI: 10.1002/adma.202310856] [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: 10/17/2023] [Revised: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Tissue ablation techniques have emerged as a critical component of modern medical practice and biomedical research, offering versatile solutions for treating various diseases and disorders. Percutaneous ablation is minimally invasive and offers numerous advantages over traditional surgery, such as shorter recovery times, reduced hospital stays, and decreased healthcare costs. Intra-procedural imaging during ablation also allows precise visualization of the treated tissue while minimizing injury to the surrounding normal tissues, reducing the risk of complications. Here, the mechanisms of tissue ablation and innovative energy delivery systems are explored, highlighting recent advancements that have reshaped the landscape of clinical practice. Current clinical challenges related to tissue ablation are also discussed, underlining unmet clinical needs for more advanced material-based approaches to improve the delivery of energy and pharmacology-based therapeutics.
Collapse
Affiliation(s)
- Hyeongseop Keum
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Enes Cevik
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Jinjoo Kim
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Yusuf M Demirlenk
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Dila Atar
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Gia Saini
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Rahul A Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Amy R Deipolyi
- Interventional Radiology, Department of Surgery, West Virginia University, Charleston Area Medical Center, Charleston, WV 25304, USA
| | - Rahmi Oklu
- Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
- Division of Vascular & Interventional Radiology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, Arizona 85054, USA
| |
Collapse
|
3
|
Morani DO, Patil PO. Review on Multifunctional Nanotherapeutics for Drug Delivery, Tumor
Imaging, and Selective Tumor Targeting by Hyaluronic Acid Coupled
Graphene Quantum Dots. CURRENT NANOSCIENCE 2024; 20:89-108. [DOI: 10.2174/1573413719666230210122445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/11/2022] [Accepted: 11/23/2022] [Indexed: 01/06/2025]
Abstract
Abstract:
Cancer is one of the most widespread life-threatening diseases, and among different
types of cancers, breast cancer is the major disease affecting many women worldwide.
Background:
Conventional chemotherapy using anticancer drugs has many drawbacks, like
poor water solubility, poor bioavailability, rapid relapse, non-specific selectivity, effect on normal
tissues, and rapid drug resistance. Thus, over the last few years, immense efforts have been
made to fabricate nanotherapeutics that will release drugs in response to stimuli.
Objective:
Nanotherapeutics based on graphene quantum dots have been acknowledged with
much gratitude in the bioscience field and investigation applications because of their distinguishing
chemical and physical properties, such as medicine delivery, biosensors, and bioimaging for
the advancement invention of disease.
Conclusion:
This paper analyzes the potential applications of graphene quantum dots for the
modified and desired release of antitumor drugs. Also, it shows graphene quantum dots' capability
to functionalize in the companionship of hyaluronic acid that operates regarding cancer cell
directing matrix in bioimaging and multimodal therapy.
Collapse
Affiliation(s)
- Dilip O. Morani
- Department of Pharmaceutics , Shri D. D. Vispute College of Pharmacy & Research Center, Devad - Vichumbe,
Panvel, Navi Mumbai-410206, India
| | - Pravin O. Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical
Education and Research, Shirpur, Dist. Dhule. 425405, India
| |
Collapse
|
4
|
Li H, Zhou Y, Guo X, Zhang Q, Ding X. The effects of irreversible electroporation triggering anti-tumor immunity and the value of its combination with immunotherapy. J Interv Med 2023; 6:107-110. [PMID: 37846332 PMCID: PMC10577062 DOI: 10.1016/j.jimed.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 10/18/2023] Open
Abstract
Recently, interventional ablation techniques have gained prominence in tumor treatment guidelines and complement traditional approaches, such as surgery, chemotherapy, and radiotherapy. Conventional ablation techniques, such as microwave, radiofrequency, and cryoablation, have been used; however, they have certain limitations, including the risk of damaging surrounding normal tissues and the heat sink effect caused by tumor blood flow.1 Irreversible electroporation (IRE), an ablation technology independent of thermal energy, is a promising alternative.2 Clinical studies have demonstrated IRE's efficacy in treating tumors, such as pancreatic and liver tumors.3 Recent research has shown that IRE can elicit specific anti-tumor immune responses in the body.5 IRE also plays a crucial role in eliminating residual tumor cells postoperatively and preventing tumor recurrence.
Collapse
Affiliation(s)
- Hengyu Li
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Yu Zhou
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoxia Guo
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Qiwei Zhang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoyi Ding
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| |
Collapse
|
5
|
Mousavi SM, Hashemi SA, Fallahi Nezhad F, Binazadeh M, Dehdashtijahromi M, Omidifar N, Ghahramani Y, Lai CW, Chiang WH, Gholami A. Innovative Metal-Organic Frameworks for Targeted Oral Cancer Therapy: A Review. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4685. [PMID: 37444999 DOI: 10.3390/ma16134685] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/12/2023] [Accepted: 05/29/2023] [Indexed: 07/15/2023]
Abstract
Metal-organic frameworks (MOFs) have proven to be very effective carriers for drug delivery in various biological applications. In recent years, the development of hybrid nanostructures has made significant progress, including developing an innovative MOF-loaded nanocomposite with a highly porous structure and low toxicity that can be used to fabricate core-shell nanocomposites by combining complementary materials. This review study discusses using MOF materials in cancer treatment, imaging, and antibacterial effects, focusing on oral cancer cells. For patients with oral cancer, we offer a regular program for accurately designing and producing various anticancer and antibacterial agents to achieve maximum effectiveness and the lowest side effects. Also, we want to ensure that the anticancer agent works optimally and has as few side effects as possible before it is tested in vitro and in vivo. It is also essential that new anticancer drugs for cancer treatment are tested for efficacy and safety before they go into further research.
Collapse
Affiliation(s)
- Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Seyyed Alireza Hashemi
- Nanomaterials and Polymer Nanocomposites Laboratory, School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Fatemeh Fallahi Nezhad
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71439-14693, Iran
| | - Mojtaba Binazadeh
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz 71557-13876, Iran
| | - Milad Dehdashtijahromi
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz 71557-13876, Iran
| | - Navid Omidifar
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz 71439-14693, Iran
| | - Yasamin Ghahramani
- Associate Professor of Endodontics Department of Endodontics, School of Dentistry Oral and Dental Disease Research Center Shiraz University of Medical Sciences, Shiraz 71956-15787, Iran
| | - Chin Wei Lai
- Nanotechnology & Catalysis Research Centre (NANOCAT), University of Malaya (UM), Kuala Lumpur 50603, Malaysia
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71439-14693, Iran
| |
Collapse
|
6
|
Tanaka HY, Nakazawa T, Enomoto A, Masamune A, Kano MR. Therapeutic Strategies to Overcome Fibrotic Barriers to Nanomedicine in the Pancreatic Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15030724. [PMID: 36765684 PMCID: PMC9913712 DOI: 10.3390/cancers15030724] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Pancreatic cancer is notorious for its dismal prognosis. The enhanced permeability and retention (EPR) effect theory posits that nanomedicines (therapeutics in the size range of approximately 10-200 nm) selectively accumulate in tumors. Nanomedicine has thus been suggested to be the "magic bullet"-both effective and safe-to treat pancreatic cancer. However, the densely fibrotic tumor microenvironment of pancreatic cancer impedes nanomedicine delivery. The EPR effect is thus insufficient to achieve a significant therapeutic effect. Intratumoral fibrosis is chiefly driven by aberrantly activated fibroblasts and the extracellular matrix (ECM) components secreted. Fibroblast and ECM abnormalities offer various potential targets for therapeutic intervention. In this review, we detail the diverse strategies being tested to overcome the fibrotic barriers to nanomedicine in pancreatic cancer. Strategies that target the fibrotic tissue/process are discussed first, which are followed by strategies to optimize nanomedicine design. We provide an overview of how a deeper understanding, increasingly at single-cell resolution, of fibroblast biology is revealing the complex role of the fibrotic stroma in pancreatic cancer pathogenesis and consider the therapeutic implications. Finally, we discuss critical gaps in our understanding and how we might better formulate strategies to successfully overcome the fibrotic barriers in pancreatic cancer.
Collapse
Affiliation(s)
- Hiroyoshi Y. Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Takuya Nakazawa
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya-shi 466-8550, Aichi, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai-shi 980-8574, Miyagi, Japan
| | - Mitsunobu R. Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
- Correspondence:
| |
Collapse
|
7
|
Bai X, Smith ZL, Wang Y, Butterworth S, Tirella A. Sustained Drug Release from Smart Nanoparticles in Cancer Therapy: A Comprehensive Review. MICROMACHINES 2022; 13:mi13101623. [PMID: 36295976 PMCID: PMC9611581 DOI: 10.3390/mi13101623] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 05/14/2023]
Abstract
Although nanomedicine has been highly investigated for cancer treatment over the past decades, only a few nanomedicines are currently approved and in the market; making this field poorly represented in clinical applications. Key research gaps that require optimization to successfully translate the use of nanomedicines have been identified, but not addressed; among these, the lack of control of the release pattern of therapeutics is the most important. To solve these issues with currently used nanomedicines (e.g., burst release, systemic release), different strategies for the design and manufacturing of nanomedicines allowing for better control over the therapeutic release, are currently being investigated. The inclusion of stimuli-responsive properties and prolonged drug release have been identified as effective approaches to include in nanomedicine, and are discussed in this paper. Recently, smart sustained release nanoparticles have been successfully designed to safely and efficiently deliver therapeutics with different kinetic profiles, making them promising for many drug delivery applications and in specific for cancer treatment. In this review, the state-of-the-art of smart sustained release nanoparticles is discussed, focusing on the design strategies and performances of polymeric nanotechnologies. A complete list of nanomedicines currently tested in clinical trials and approved nanomedicines for cancer treatment is presented, critically discussing advantages and limitations with respect to the newly developed nanotechnologies and manufacturing methods. By the presented discussion and the highlight of nanomedicine design criteria and current limitations, this review paper could be of high interest to identify key features for the design of release-controlled nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xue Bai
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Zara L. Smith
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Yuheng Wang
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Sam Butterworth
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Annalisa Tirella
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- BIOtech-Center for Biomedical Technologies, Department of Industrial Engineering, University of Trento, Via delle Regole 101, 38123 Trento, Italy
- Correspondence:
| |
Collapse
|
8
|
Mousavi SM, Hashemi SA, Ghahramani Y, Azhdari R, Yousefi K, Gholami A, Fallahi Nezhad F, Vijayakameswara Rao N, Omidifar N, Chiang WH. Antiproliferative and Apoptotic Effects of Graphene Oxide @AlFu MOF Based Saponin Natural Product on OSCC Line. Pharmaceuticals (Basel) 2022; 15:ph15091137. [PMID: 36145358 PMCID: PMC9504826 DOI: 10.3390/ph15091137] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 12/16/2022] Open
Abstract
The increasing rate of oral squamous cell carcinoma (OSCC) and the undesirable side effects of anticancer agents have enhanced the demand for the development of efficient, detectable, and targeted anticancer systems. Saponins are a diverse family of natural glycosides that have recently been evaluated as an effective compound for the targeted therapy of squamous cell carcinoma. Due to their porous nature and stable structure, metal–organic frameworks (MOFs) are a well-known substance form for various biological applications, such as drug delivery. In this study, we fabricated a novel hybrid, highly porous and low-toxic saponin-loaded nanostructure by modifying graphene oxide (GO)/reduced GO (rGO) with aluminum fumarate (AlFu) as MOF core–shell nanocomposite. The characterization of the nanostructures was investigated by FTIR, TEM, EDX, FESEM, and BET. MTT assay was used to investigate the anticancer activity of these compounds on OSCC and PDL normal dental cells. The effect of the nanocomposites on OSCC was then investigated by studying apoptosis and necrosis using flow cytometry. The GO/rGO was decorated with a saponin–AlFu mixture to further investigate cytotoxicity. The results of the MTT assay showed that PDL cells treated with AlFu–GO–saponin at a concentration of 250 μg/mL had a viability of 74.46 ± 16.02%, while OSCC cells treated with this sample at a similar concentration had a viability of only 38.35 ± 19.9%. The anticancer effect of this nanostructure on OSCC was clearly demonstrated. Moreover, the number of apoptotic cells in the AlFu–GO–saponin and AlFu–rGO–saponin groups was 10.98 ± 2.36%–26.90 ± 3.24% and 15.9 ± 4.08%–29.88 ± 0.41%, respectively, compared with 2.52 ± 0.78%–1.31 ± 0.62% in the untreated group. This significant increase in apoptotic effect observed with AlFu–rGO–saponin was also reflected in the significant anticancer effect of saponin-loaded nanostructures. Therefore, this study suggests that an effective saponin delivery system protocol for the precise design and fabrication of anticancer nanostructures for OSCC therapy should be performed prior to in vivo evaluations.
Collapse
Affiliation(s)
- Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 106335, Taiwan
| | - Seyyed Alireza Hashemi
- Nanomaterials and Polymer Nanocomposites Laboratory, School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Yasmin Ghahramani
- Department of Endodontics, Shiraz University of Medical Sciences, Shiraz 71956-15787, Iran
| | - Rouhollah Azhdari
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Khadijeh Yousefi
- Department of Dental Materials and Biomaterials Research Centre, Shiraz Dental School, Shiraz University of Medical Sciences, Shiraz 71956-15787, Iran
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Fatemeh Fallahi Nezhad
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Neralla Vijayakameswara Rao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 106335, Taiwan
| | - Navid Omidifar
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 106335, Taiwan
| |
Collapse
|
9
|
Tang X, Du X, Yu Y, Qin M, Qian L, Zhang M, Yang Y, Yu Q, Gan Z. Deep-Penetrating Triple-Responsive Prodrug Nanosensitizer Actuates Efficient Chemoradiotherapy in Pancreatic Ductal Adenocarcinoma Models. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202834. [PMID: 35808966 DOI: 10.1002/smll.202202834] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Chemoradiotherapy (CRT) is the most accepted treatment for locally advanced pancreatic ductal adenocarcinoma (PDAC) and can significantly improve the R0 resection rate. However, there are few long-term survivors after CRT. Although some polymer nanoparticles have shown potential in alleviating the dose-limiting toxicity and assisting the chemotherapy of PDAC, there are few efficient nanosensitizers (NS) available for CRT of this malignancy, especially in the context of its hypoxic nature. Herein, based on the biological features of PDAC, a γ-glutamyl transpeptidase (GGT)/glutathione (GSH)/hypoxia triple-responsive prodrug NS to overcome the biological barrier and microenvironmental limitations confronted by CRT in PDAC is developed. Due to triple-responsiveness, deep tumor penetration, GSH/hypoxia-responsive drug release/activation, and hypoxia-induced chemoradio-sensitization can be simultaneously achieved with this NS. As a result, tumor shrinkage after CRT with this NS can be observed in both subcutaneous and orthotopic PDAC models, foreshadowing its potential in clinical neoadjuvant CRT.
Collapse
Affiliation(s)
- Xiaohu Tang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xiaomeng Du
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, P. R. China
| | - Yanting Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Meng Qin
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Lili Qian
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Meng Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yan Yang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Qingsong Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Zhihua Gan
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
10
|
Nanotheranostics for Image-Guided Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14050917. [PMID: 35631503 PMCID: PMC9144228 DOI: 10.3390/pharmaceutics14050917] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Image-guided nanotheranostics have the potential to represent a new paradigm in the treatment of cancer. Recent developments in modern imaging and nanoparticle design offer an answer to many of the issues associated with conventional chemotherapy, including their indiscriminate side effects and susceptibility to drug resistance. Imaging is one of the tools best poised to enable tailoring of cancer therapies. The field of image-guided nanotheranostics has the potential to harness the precision of modern imaging techniques and use this to direct, dictate, and follow site-specific drug delivery, all of which can be used to further tailor cancer therapies on both the individual and population level. The use of image-guided drug delivery has exploded in preclinical and clinical trials although the clinical translation is incipient. This review will focus on traditional mechanisms of targeted drug delivery in cancer, including the use of molecular targeting, as well as the foundations of designing nanotheranostics, with a focus on current clinical applications of nanotheranostics in cancer. A variety of specially engineered and targeted drug carriers, along with strategies of labeling nanoparticles to endow detectability in different imaging modalities will be reviewed. It will also introduce newer concepts of image-guided drug delivery, which may circumvent many of the issues seen with other techniques. Finally, we will review the current barriers to clinical translation of image-guided nanotheranostics and how these may be overcome.
Collapse
|
11
|
Chen L, Jia L, Tian Z, Yang Y, Zhao K. Elderly Patients with Nondistant Metastatic Pancreatic Head Adenocarcinoma Cannot Benefit from More Radical Surgery. Int J Endocrinol 2022; 2022:6469740. [PMID: 35479664 PMCID: PMC9038409 DOI: 10.1155/2022/6469740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/27/2022] [Accepted: 04/01/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The incidence of pancreatic cancer continues to rise globally, with pancreatic head cancer accounting for nearly 60-70%. Pancreatic head cancer occurs mainly in people over the age of 60, and its morbidity and mortality increase with age. We investigated whether these elderly patients with nondistant metastases would benefit more from expanded pancreaticoduodenectomy (EPD) compared with standard pancreaticoduodenectomy (SPD). METHODS 3317 elderly patients with pancreatic head cancer from the SEER database were included in the study based on the inclusion and exclusion criteria. These patients were divided into a nonsurgical group and surgical group (including EPD and SPD). Univariate and multivariate Cox proportional hazards models were applied to identify the independent risk factors for cancer-specific survival (CSS). The survival differences between the nonsurgical group and surgical group were compared. Propensity score matching (PSM) methods were applied to balance covariates and reduce the interference of confounding variables. The two groups of patients were matched in a 1 : 1 ratio, and the covariates between the two groups were compared to verify the matching validity. The survival difference in different groups was compared after the matching analysis. RESULTS 3317 enrolled patients were divided into the surgical group (n = 984) and nonsurgical group (n = 2333). Before PSM, there were significant differences in overall survival (OS) and CSS between the nonsurgical group and surgical group (median OS: 8 months vs. 20 months, P < 0.001; median CSS: 8 months vs. 22 months, P < 0.001). The multivariate CSS Cox regression analysis demonstrated surgery is an independent risk factor. However, no significant differences were founded between the SPD and EPD groups (median OS: 20 months vs. 22 months, P=0.636; median CSS: 22 months vs. 22 months, P=0.270). After PSM, there were also no significant differences in OS and CSS between the SPD and EPD groups (median OS: 23 months vs. 18 months, P=0.415; median CSS: 26 months vs. 18 months, P=0.329). CONCLUSION This study uses PSM to evaluate the effects of EPD and SPD for elderly patients with nondistant metastatic pancreatic head adenocarcinoma. It found that surgery is an independent prognostic factor, but expanded surgery has no survival advantage for these patients, whereas SPD provides a better survival advantage than EPD. SPD is a reasonable treatment option for these patients.
Collapse
Affiliation(s)
- Li Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Lanning Jia
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Zhigang Tian
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Yang Yang
- Department of Anorectal Surgery, Anorectal Surgery Center, Union's Hospital of Tianjin, Tianjin, China
| | - Ke Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Nanomedicine in Pancreatic Cancer: Current Status and Future Opportunities for Overcoming Therapy Resistance. Cancers (Basel) 2021; 13:cancers13246175. [PMID: 34944794 PMCID: PMC8699181 DOI: 10.3390/cancers13246175] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Despite access to a vast arsenal of anticancer agents, many fail to realise their full therapeutic potential in clinical practice. One key determinant of this is the evolution of multifaceted resistance mechanisms within the tumour that may either pre-exist or develop during the course of therapy. This is particularly evident in pancreatic cancer, where limited responses to treatment underlie dismal survival rates, highlighting the urgent need for new therapeutic approaches. Here, we discuss the major features of pancreatic tumours that contribute to therapy resistance, and how they may be alleviated through exploitation of the mounting and exciting promise of nanomedicines; a unique collection of nanoscale platforms with tunable and multifunctional capabilities that have already elicited a widespread impact on cancer management. Abstract The development of drug resistance remains one of the greatest clinical oncology challenges that can radically dampen the prospect of achieving complete and durable tumour control. Efforts to mitigate drug resistance are therefore of utmost importance, and nanotechnology is rapidly emerging for its potential to overcome such issues. Studies have showcased the ability of nanomedicines to bypass drug efflux pumps, counteract immune suppression, serve as radioenhancers, correct metabolic disturbances and elicit numerous other effects that collectively alleviate various mechanisms of tumour resistance. Much of this progress can be attributed to the remarkable benefits that nanoparticles offer as drug delivery vehicles, such as improvements in pharmacokinetics, protection against degradation and spatiotemporally controlled release kinetics. These attributes provide scope for precision targeting of drugs to tumours that can enhance sensitivity to treatment and have formed the basis for the successful clinical translation of multiple nanoformulations to date. In this review, we focus on the longstanding reputation of pancreatic cancer as one of the most difficult-to-treat malignancies where resistance plays a dominant role in therapy failure. We outline the mechanisms that contribute to the treatment-refractory nature of these tumours, and how they may be effectively addressed by harnessing the unique capabilities of nanomedicines. Moreover, we include a brief perspective on the likely future direction of nanotechnology in pancreatic cancer, discussing how efforts to develop multidrug formulations will guide the field further towards a therapeutic solution for these highly intractable tumours.
Collapse
|
13
|
Sujai PT, Shamjith S, Joseph MM, Maiti KK. Elucidating Gold-MnO 2 Core-Shell Nanoenvelope for Real Time SERS-Guided Photothermal Therapy on Pancreatic Cancer Cells. ACS APPLIED BIO MATERIALS 2021; 4:4962-4972. [PMID: 35007044 DOI: 10.1021/acsabm.1c00241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pancreatic cancer represents one of the most aggressive in nature with a miserable prognosis that warrants efficient diagnostic and therapeutic interventions. Herein, a MnO2 overlaid gold nanoparticle (AuNPs) based photothermal theranostic nanoenvelope (PTTNe:MnO2@AuNPs) was fabricated to substantiate surface-enhanced Raman spectroscopy (SERS) guided real-time monitoring of photothermal therapy (PTT) in pancreatic cancer cells. A sharp enhancement of the fingerprint Raman signature of MnO2 at 569 cm-1 exhibited as a marker peak for the first time to elucidate the intracellular PTT event. In this strategic design, the leftover bare AuNPs after the degradation of the MnO2 layer from the nanoenvelope in the presence of intracellular H2O2 enabled real-time tracking of biomolecular changes of Raman spectral variations during PTT. Moreover, the surface of the as-synthesized nanoenvelope was functionalized with a pancreatic cancer cell targeting peptide sequence for cholecystokinin fashioned the PTTNe with admirable stability and biocompatibility. Finally, the precise cell death mechanism was explicitly assessed by SERS spectral analysis as a complementary technique. This targeted phototheranostic approach demonstrated in pancreatic cancer cells presented a therapeutically viable prototype for futuristic personalized cancer nanomedicine.
Collapse
Affiliation(s)
- Palasseri T Sujai
- Chemical Sciences and Technology Division (CSTD), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019 Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shanmughan Shamjith
- Chemical Sciences and Technology Division (CSTD), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019 Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manu M Joseph
- Chemical Sciences and Technology Division (CSTD), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019 Kerala, India
| | - Kaustabh Kumar Maiti
- Chemical Sciences and Technology Division (CSTD), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019 Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
14
|
Asadi S, Bianchi L, De Landro M, Korganbayev S, Schena E, Saccomandi P. Laser-induced optothermal response of gold nanoparticles: From a physical viewpoint to cancer treatment application. JOURNAL OF BIOPHOTONICS 2021; 14:e202000161. [PMID: 32761778 DOI: 10.1002/jbio.202000161] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/15/2020] [Accepted: 07/28/2020] [Indexed: 06/11/2023]
Abstract
Gold nanoparticles (GNPs)-based photothermal therapy (PTT) is a promising minimally invasive thermal therapy for the treatment of focal malignancies. Although GNPs-based PTT has been known for over two decades and GNPs possess unique properties as therapeutic agents, the delivery of a safe and effective therapy is still an open question. This review aims at providing relevant and recent information on the usage of GNPs in combination with the laser to treat cancers, pointing out the practical aspects that bear on the therapy outcome. Emphasis is given to the assessment of the GNPs' properties and the physical mechanisms underlying the laser-induced heat generation in GNPs-loaded tissues. The main techniques available for temperature measurement and the current theoretical simulation approaches predicting the therapeutic outcome are reviewed. Topical challenges in delivering safe thermal dosage are also presented with the aim to discuss the state-of-the-art and the future perspective in the field of GNPs-mediated PTT.
Collapse
Affiliation(s)
- Somayeh Asadi
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| | - Leonardo Bianchi
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| | - Martina De Landro
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| | | | - Emiliano Schena
- Laboratory of Measurement and Biomedical Instrumentation, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Paola Saccomandi
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
15
|
Si K, Xue Y, Yu X, Zhu X, Li Q, Gong W, Liang T, Duan S. Fully end-to-end deep-learning-based diagnosis of pancreatic tumors. Am J Cancer Res 2021; 11:1982-1990. [PMID: 33408793 PMCID: PMC7778580 DOI: 10.7150/thno.52508] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Artificial intelligence can facilitate clinical decision making by considering massive amounts of medical imaging data. Various algorithms have been implemented for different clinical applications. Accurate diagnosis and treatment require reliable and interpretable data. For pancreatic tumor diagnosis, only 58.5% of images from the First Affiliated Hospital and the Second Affiliated Hospital, Zhejiang University School of Medicine are used, increasing labor and time costs to manually filter out images not directly used by the diagnostic model. Methods: This study used a training dataset of 143,945 dynamic contrast-enhanced CT images of the abdomen from 319 patients. The proposed model contained four stages: image screening, pancreas location, pancreas segmentation, and pancreatic tumor diagnosis. Results: We established a fully end-to-end deep-learning model for diagnosing pancreatic tumors and proposing treatment. The model considers original abdominal CT images without any manual preprocessing. Our artificial-intelligence-based system achieved an area under the curve of 0.871 and a F1 score of 88.5% using an independent testing dataset containing 107,036 clinical CT images from 347 patients. The average accuracy for all tumor types was 82.7%, and the independent accuracies of identifying intraductal papillary mucinous neoplasm and pancreatic ductal adenocarcinoma were 100% and 87.6%, respectively. The average test time per patient was 18.6 s, compared with at least 8 min for manual reviewing. Furthermore, the model provided a transparent and interpretable diagnosis by producing saliency maps highlighting the regions relevant to its decision. Conclusions: The proposed model can potentially deliver efficient and accurate preoperative diagnoses that could aid the surgical management of pancreatic tumor.
Collapse
|
16
|
Yang Y, Chen P, Zhao L, Zhang B, Xu C, Zhang H, Zhou J. Design, synthesis and biological evaluation of imidazolopyridone derivatives as novel BRD4 inhibitors. Bioorg Med Chem 2020; 29:115857. [PMID: 33191086 DOI: 10.1016/j.bmc.2020.115857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 11/17/2022]
Abstract
Bromodomain containing protein 4 (BRD4) has been demonstrated to play critical roles in cellular proliferation and cell cycle progression. In this study, using the BRD4 inhibitor Fragment 9 as a lead compound, a series of imidazolopyridone derivatives were designed and tested for their inhibitory activity against BRD4 protein in vitro. Among them, HB100-A7 showed excellent BRD4(1) inhibitory activities with an IC50 value of 0.035 μM in amplified luminescent proximity homogeneous assay (Alphascreen). The result of MTT assay showed that HB100-A7 could suppress the proliferation of pancreatic cancer cells. In addition, flow cytometry further illustrated that HB100-A7 treatment resulted in G0/G1 phase arrest and promoted apoptosis of BxPc3 cells. Furthermore, the in vivo study found that HB100-A7 displayed significant tumor growth inhibition in a pancreatic mouse tumor model (Panc-02). Moreover, IHC staining suggested that HB100-A7 induce cell apoptosis in pancreatic cancer tumor tissue. Together, this study revealed, for the first time, HB100-A7 is a promising lead compound for further development as a new generation of small molecule inhibitors targeting the BRD4 protein.
Collapse
Affiliation(s)
- Yifei Yang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Pan Chen
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Leilei Zhao
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bing Zhang
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China
| | - Changliang Xu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China.
| | - Huibin Zhang
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
17
|
Wang G, Wu B, Li Q, Chen S, Jin X, Liu Y, Zhou Z, Shen Y, Huang P. Active Transportation of Liposome Enhances Tumor Accumulation, Penetration, and Therapeutic Efficacy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004172. [PMID: 33030305 DOI: 10.1002/smll.202004172] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/04/2020] [Indexed: 06/11/2023]
Abstract
Liposomes are the first and mostly explored nanocarriers for cancer drug delivery, which have shown great promise in clinical applications, but their limited accumulation and penetration into the tumor interstitial space, significantly reduce the therapeutic efficacy. Here, a γ-glutamyltranspeptidase (GGT)-triggered charge-switchable approach is reported that can trigger the fast endocytosis and transcytosis of the liposome in tumor microenvironments to overcome the harsh biological barriers in tumor tissues. The active transporting liposomal nanocarrier (GCSDL) is prepared by surface modification with a glutathione (GSH) moiety and encapsulated with doxorubicin (DOX). When the GCSDL contacts with tumor vascular endothelial cells, the overexpressed GGT enzyme on cytomembrane catalyzes the hydrolysis of GSH to generate cationic primary amines. The cationic GCSDL triggers fast caveolae-mediated endocytosis and vesicle-mediated transcytosis, resulting in sequential transcytosis to augment its tumor accumulation and penetration. Along with continual intercellular transportation, GCSDL can release DOX throughout the tumor to induce cancer cell apoptosis, resulting in complete eradication of hepatocellular carcinoma and cessation of pancreatic ductal adenocarcinoma's progression. This study develops an efficient strategy to realize high tumor accumulation and deep penetration for the liposomal drug delivery system via active transcytosis.
Collapse
Affiliation(s)
- Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Bihan Wu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qunying Li
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Siqin Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiaoqin Jin
- Department of Pathology, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yajing Liu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhuxian Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| | - Youqing Shen
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
18
|
Herrmann K, Schwaiger M, Lewis JS, Solomon SB, McNeil BJ, Baumann M, Gambhir SS, Hricak H, Weissleder R. Radiotheranostics: a roadmap for future development. Lancet Oncol 2020; 21:e146-e156. [PMID: 32135118 DOI: 10.1016/s1470-2045(19)30821-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/25/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Radiotheranostics, injectable radiopharmaceuticals with antitumour effects, have seen rapid development over the past decade. Although some formulations are already approved for human use, more radiopharmaceuticals will enter clinical practice in the next 5 years, potentially introducing new therapeutic choices for patients. Despite these advances, several challenges remain, including logistics, supply chain, regulatory issues, and education and training. By highlighting active developments in the field, this Review aims to alert practitioners to the value of radiotheranostics and to outline a roadmap for future development. Multidisciplinary approaches in clinical trial design and therapeutic administration will become essential to the continued progress of this evolving therapeutic approach.
Collapse
Affiliation(s)
- Ken Herrmann
- Clinic for Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Stephen B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Barbara J McNeil
- Department of Radiology, Brigham and Women's Hospital, and Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | | | - Sanjiv S Gambhir
- Department of Radiology and Molecular Imaging Program, Stanford University, Stanford, CA, USA
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ralph Weissleder
- Department of Radiology, and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Huang X, Wang M, You Q, Kong J, Zhang H, Yu C, Wang Y, Wang H, Huang R. Synthesis, mechanisms of action, and toxicity of novel aminophosphonates derivatives conjugated irinotecan in vitro and in vivo as potent antitumor agents. Eur J Med Chem 2020; 189:112067. [PMID: 31972391 DOI: 10.1016/j.ejmech.2020.112067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 01/17/2023]
Abstract
Twenty novel aminophosphonates derivatives (5a-5j and 6a-6j) conjugated irinotecan were synthesized through esterification reaction, and evaluated their anticancer activities using MTT assay. In vitro evaluation revealed that they displayed similar or superior cytotoxicity compared to the positive drug irinotecan against A549, MCF-7, SK-OV-3, MG-63, U2OS and multidrug-resistant (MDR) SK-OV-3/CDDP cancer cell lines. Among them, 9b displayed the most potent activity, with IC50 values of 0.92-3.23 μM against five human cancer cells, which exhibited a 5.4-19.1-fold increase in activity compared to the reference drug irinotecan, respectively. Moreover, cellular mechanism studies suggested that 9b arrested cell cycle at S stage and induced cell apoptosis along with the decrease of mitochondrial membrane potential (MMP). Interestingly, 9b significantly inhibited tumor growth in SK-OV-3 xenograft models in vivo without apparent toxicity, which was better than the positive drug irinotecan. Taken together, 9b possessed potent antitumor activity and may be a promising candidate for the potential treatment of human ovarian cancer cells.
Collapse
Affiliation(s)
- Xiaochao Huang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China; College of Biotechnology, Guilin Medical University, Guilin, 541004, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| | - Meng Wang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Qinghong You
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Jing Kong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Haijiang Zhang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Chunhao Yu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Yanming Wang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| | - Rizhen Huang
- College of Biotechnology, Guilin Medical University, Guilin, 541004, China.
| |
Collapse
|
20
|
Iannazzo D, Pistone A, Celesti C, Triolo C, Patané S, Giofré SV, Romeo R, Ziccarelli I, Mancuso R, Gabriele B, Visalli G, Facciolà A, Di Pietro A. A Smart Nanovector for Cancer Targeted Drug Delivery Based on Graphene Quantum Dots. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E282. [PMID: 30781623 PMCID: PMC6409783 DOI: 10.3390/nano9020282] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/08/2019] [Accepted: 02/10/2019] [Indexed: 12/12/2022]
Abstract
Graphene quantum dots (GQD), the new generation members of graphene-family, have shown promising applications in anticancer therapy. In this study, we report the synthesis of a fluorescent and biocompatible nanovector, based on GQD, for the targeted delivery of an anticancer drug with benzofuran structure (BFG) and bearing the targeting ligand riboflavin (RF, vitamin B2). The highly water-dispersible nanoparticles, synthesized from multi-walled carbon nanotubes (MWCNT) by prolonged acidic treatment, were linked covalently to the drug by means of a cleavable PEG linker while the targeting ligand RF was conjugated to the GQD by π⁻π interaction using a pyrene linker. The cytotoxic effect of the synthesized drug delivery system (DDS) GQD-PEG-BFG@Pyr-RF was tested on three cancer cell lines and this effect was compared with that exerted by the same nanovector lacking the RF ligand (GQD-PEG-BFG) or the anticancer drug (GQD@Pyr-RF). The results of biological tests underlined the low cytotoxicity of the GQD sample and the cytotoxic activity of the DDS against the investigated cancer cell lines with a higher or similar potency to that exerted by the BFG alone, thus opening new possibilities for the use of this drug or other anticancer agents endowed of cytotoxicity and serious side effects.
Collapse
Affiliation(s)
- Daniela Iannazzo
- Department of Engineering, University of Messina, Contrada Di Dio, I-98166 Messina, Italy.
| | - Alessandro Pistone
- Department of Engineering, University of Messina, Contrada Di Dio, I-98166 Messina, Italy.
| | - Consuelo Celesti
- Department of Engineering, University of Messina, Contrada Di Dio, I-98166 Messina, Italy.
| | - Claudia Triolo
- Department of Mathematical and Computer Sciences, Physical Sciences and Earth Sciences, Viale F. Stagno d'Alcontres, 98166 Messina, Italy.
| | - Salvatore Patané
- Department of Mathematical and Computer Sciences, Physical Sciences and Earth Sciences, Viale F. Stagno d'Alcontres, 98166 Messina, Italy.
| | - Salvatore V Giofré
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, I-98168 Messina, Italy.
| | - Roberto Romeo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, I-98168 Messina, Italy.
| | - Ida Ziccarelli
- Laboratory of Industrial and Synthetic Organic Chemistry (LISOC), Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 12/C, 87036 Arcavacata di Rende (CS), Italy.
| | - Raffaella Mancuso
- Laboratory of Industrial and Synthetic Organic Chemistry (LISOC), Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 12/C, 87036 Arcavacata di Rende (CS), Italy.
| | - Bartolo Gabriele
- Laboratory of Industrial and Synthetic Organic Chemistry (LISOC), Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 12/C, 87036 Arcavacata di Rende (CS), Italy.
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphological and Functional Images, University Hospital of Messina, Via Consolare Valeria, 1, 98100 Messina, Italy.
| | - Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphological and Functional Images, University Hospital of Messina, Via Consolare Valeria, 1, 98100 Messina, Italy.
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphological and Functional Images, University Hospital of Messina, Via Consolare Valeria, 1, 98100 Messina, Italy.
| |
Collapse
|
21
|
Rhein sensitizes human pancreatic cancer cells to EGFR inhibitors by inhibiting STAT3 pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:31. [PMID: 30674340 PMCID: PMC6343257 DOI: 10.1186/s13046-018-1015-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 12/19/2018] [Indexed: 01/05/2023]
Abstract
Background Rhein is a lipophilic anthraquinone extensively found in medicinal herbs. Emerging evidence suggests that rhein has significant antitumor effects, supporting its potential use as an antitumor agent. The IL6/STAT3 signaling pathway has been suggested as an attractive target for the discovery of novel cancer therapeutics. Methods The human pancreatic cancer cell lines AsPC-1, Patu8988T, BxPC-3 and PANC-1, and immunodeficient mice were chosen as models to study the effects of rhein. The potent antiproliferative and proapoptotic effects of rhein were examined by cell viability, cellular morphology, apoptosis and colony formation assays. The STAT3 luciferase report assay, immunostaining analysis and Western blot analysis revealed the inhibition of the IL6/STAT3 signaling axis. Results Apoptosis was induced by adjunctive use of rhein with epidermal growth factor receptor (EGFR) inhibitors in pancreatic cancer cells as verified by cell apoptosis analysis and changes in the expression level of apoptotic/anti-apoptotic proteins BCL-2, BAX, Caspase 3 and Cl-PARP. Suppression of the phosphorylation of STAT3 and EGFR were also observed as a result of the treatment with a combination of rhein and EGFR inhibitors. Most interestingly, it was found that rhein considerably sensitized cells to erlotinib, thus suppressing tumor growth in PANC-1 and BxPC-3 xenograft models. The in vivo anti-tumor effect was associated with increased apoptosis and combined inhibition of the STAT3 and EGFR pathways in tumor remnants. Conclusions Rhein sensitizes human pancreatic cancer cells to EGFR inhibitors through inhibition of STAT3. Taken together, the results indicate that rhein offers a novel blueprint for pancreatic cancer therapy, particularly when combined with EGFR inhibitors. Electronic supplementary material The online version of this article (10.1186/s13046-018-1015-9) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Liu X, Jiang J, Chan R, Ji Y, Lu J, Liao YP, Okene M, Lin J, Lin P, Chang CH, Wang X, Tang I, Zheng E, Qiu W, Wainberg ZA, Nel AE, Meng H. Improved Efficacy and Reduced Toxicity Using a Custom-Designed Irinotecan-Delivering Silicasome for Orthotopic Colon Cancer. ACS NANO 2019; 13:38-53. [PMID: 30525443 PMCID: PMC6554030 DOI: 10.1021/acsnano.8b06164] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Irinotecan is a key chemotherapeutic agent for the treatment of colorectal (CRC) and pancreatic (PDAC) cancer. Because of a high incidence of bone marrow and gastrointestinal (GI) toxicity, Onivyde (a liposome) was introduced to provide encapsulated irinotecan (Ir) delivery in PDAC patients. While there is an ongoing clinical trial (NCT02551991) to investigate the use of Onivyde as a first-line option to replace irinotecan in FOLFIRINOX, the liposomal formulation is currently prescribed as a second-line treatment option (in combination with 5-fluorouracil and leucovorin) for patients with metastatic PDAC who failed gemcitabine therapy. However, the toxicity of Onivyde remains a concern that needs to be addressed for use in CRC as well. Our goal was to custom design a mesoporous silica nanoparticle (MSNP) carrier for encapsulated irinotecan delivery in a robust CRC model. This was achieved by developing an orthotopic tumor chunk model in immunocompetent mice. With a view to increase the production volume and to expand the disease applications, the carrier design was improved by using an ethanol exchange method for coating of a supported lipid bilayer (LB) that entraps a protonating agent. The encapsulated protonating agent was subsequently used for remote loading of irinotecan. The excellent irinotecan loading capacity and stability of the LB-coated MSNP carrier, also known as a "silicasome", previously showed improved efficacy and reduced toxicity when compared to an in-house liposomal carrier in a PDAC model. Intravenous injection of the silicasomes in a well-developed orthotopic colon cancer model in mice demonstrated improved pharmacokinetics and tumor drug content over free drug and Onivyde. Moreover, improved drug delivery was accompanied by substantially improved efficacy, increased survival, and reduced bone marrow and GI toxicity compared to the free drug and Onivyde. We also confirmed that the custom-designed irinotecan silicasomes outperform Onivyde in an orthotopic PDAC model. In summary, the Ir-silicasome appears to be promising as a treatment option for CRC in humans based on improved efficacy and the carrier's favorable safety profile.
Collapse
Affiliation(s)
- Xiangsheng Liu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Ryan Chan
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Ying Ji
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Jianqin Lu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Michael Okene
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Joshua Lin
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Paulina Lin
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Chong Hyun Chang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Ivanna Tang
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Emily Zheng
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Waveley Qiu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Zev A. Wainberg
- Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Andre E. Nel
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Huan Meng
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
23
|
El-Zahaby SA, Elnaggar YSR, Abdallah OY. Reviewing two decades of nanomedicine implementations in targeted treatment and diagnosis of pancreatic cancer: An emphasis on state of art. J Control Release 2019; 293:21-35. [PMID: 30445002 DOI: 10.1016/j.jconrel.2018.11.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/10/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is nowadays the most life-threatening cancer type worldwide. The problem of poor diagnosis, anti-neoplastics resistance and biopharmaceutical drawbacks of effective anti-cancer drugs lead to worsen disease state. Nanotechnology-based carrier systems used in both imaging and treatment procedures had solved many of these problems. It is critical to develop advanced detection method to save patients from being too late diagnosed. Targeting the pancreatic cancer cells as well helped in decreasing the side effects associated with normal cells destruction. Drug resistance is another challenge in pancreatic cancer management that can be solved by thorough understanding of the microenvironment associated with the disease to design creative nanocarriers. This is the first article to review multifaceted approaches of nanomedicine in pancreatic cancer detection and management. Additionally, mortality rates in selected Arab and European countries were illustrated herein. An emphasis was given on therapeutic and diagnostic challenges and different nanotechnologies adopted to overcome. The four main approaches encompassed nanomedicine for herbal treatment, nanomedicine of synthetic anti-cancer drugs, metal nanoparticles as a distinct treatment policy and nanotechnology for cancer diagnosis. Future research perspectives have been finally proposed.
Collapse
Affiliation(s)
- Sally A El-Zahaby
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
24
|
El-Zahaby SA, Elnaggar YS, Abdallah OY. Reviewing two decades of nanomedicine implementations in targeted treatment and diagnosis of pancreatic cancer: An emphasis on state of art. J Control Release 2019. [DOI: https://doi.org/10.1016/j.jconrel.2018.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
25
|
Di Carlo C, Brandi J, Cecconi D. Pancreatic cancer stem cells: Perspectives on potential therapeutic approaches of pancreatic ductal adenocarcinoma. World J Stem Cells 2018; 10:172-182. [PMID: 30631392 PMCID: PMC6325076 DOI: 10.4252/wjsc.v10.i11.172] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/10/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive solid tumours of the pancreas, characterised by a five-year survival rate less than 8%. Recent reports that pancreatic cancer stem cells (PCSCs) contribute to the tumorigenesis, progression, and chemoresistance of pancreatic cancer have prompted the investigation of new therapeutic approaches able to directly target PCSCs. In the present paper the non-cancer related drugs that have been proposed to target CSCs that could potentially combat pancreatic cancer are reviewed and evaluated. The role of some pathways and deregulated proteins in PCSCs as new therapeutic targets are also discussed with a focus on selected specific inhibitors. Finally, advances in the development of nanoparticles for targeting PCSCs and site-specific drug delivery are highlighted, and their limitations considered.
Collapse
Affiliation(s)
- Claudia Di Carlo
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy
| | - Jessica Brandi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy.
| | - Daniela Cecconi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy
| |
Collapse
|
26
|
Huang D, Zhao D, Wang X, Li C, Yang T, Du L, Wei Z, Cheng Q, Cao H, Liang Z, Huang Y, Li Z. Efficient delivery of nucleic acid molecules into skin by combined use of microneedle roller and flexible interdigitated electroporation array. Am J Cancer Res 2018; 8:2361-2376. [PMID: 29721085 PMCID: PMC5928895 DOI: 10.7150/thno.23438] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 02/07/2018] [Indexed: 12/31/2022] Open
Abstract
Rationale: Delivery of nucleic acid molecules into skin remains a main obstacle for various types of gene therapy or vaccine applications. Here we propose a novel electroporation approach via combined use of a microneedle roller and a flexible interdigitated electroporation array (FIEA) for efficient delivery of DNA and siRNA into mouse skin. Methods: Using micromachining technology, closely spaced gold electrodes were made on a pliable parylene substrate to form a patch-like electroporation array, which enabled close surface contact between the skin and electrodes. Pre-penetration of the skin with a microneedle roller resulted in the formation of microchannels in the skin, which played a role as liquid electrodes in the skin and provided a uniform and deep electric field in the tissue when pulse stimulation was applied by FIEA. Results: Using this proposed method, gene (RFP) expression and siRNA transfection were successfully achieved in normal mice skin. Anti-SCD1 siRNA electroporated via this method mediated significant gene silencing in the skin. Moreover, electroporation assisted by the microneedle roller showed significant advantages over treatment with FIEA alone. This allowed nucleic acid transportation at low voltage, with ideal safety outcomes. Principal conclusions: Hence, the proposed electroporation approach in this study constitutes a novel way for delivering siRNA and DNA, and even other nucleic acid molecules, to mouse skin in vivo, potentially supporting clinical application in the treatment of skin diseases or intradermal/subcutaneous vaccination.
Collapse
|
27
|
Yuan Q, Zhang Y, Li J, Cao G, Yang W. High expression of microRNA-4295 contributes to cell proliferation and invasion of pancreatic ductal adenocarcinoma by the down-regulation of Glypican-5. Biochem Biophys Res Commun 2018; 497:73-79. [PMID: 29407175 DOI: 10.1016/j.bbrc.2018.02.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/03/2018] [Indexed: 12/21/2022]
Abstract
A growing amount of evidence has documented that Glypican-5 (GPC5) is an important regulator of tumor progression. However, little is known about the role of GPC5 in pancreatic ductal adenocarcinoma (PDAC). In this study, we aimed to investigate the potential function and regulatory mechanism of GPC5 in PDAC. We found that GPC5 expression was significantly down-regulated in PDAC cell lines. The overexpression of GPC5 inhibited cell proliferation and the invasion of PDAC cells. In addition, the overexpression of GPC5 suppressed Wnt/β-catenin signaling in PDAC cells. Bioinformatic analysis predicted that GPC5 was a target gene of microRNA-4295 (miR-4295). The inhibition of miR-4295 significantly up-regulated the expression of GPC5. Moreover, the inhibition of miR-4295 inhibited the proliferation, invasion and Wnt/β-catenin signaling in PDAC cells. Notably, the knockdown of GPC5 partially reversed the anti-tumor effect of miR-4295 inhibition. Taken together, our results suggest GPC5 as a tumor suppressor in PDAC and its expression is possibly regulated by miR-4295. Our study indicates that the miR-4295/GPC5 axis may play an important role in the pathogenesis of PADC and has potential applications for the development of PDAC therapy.
Collapse
Affiliation(s)
- Qinggong Yuan
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China
| | - Yan Zhang
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China
| | - Junhui Li
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China
| | - Gang Cao
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China
| | - Wenbin Yang
- Department of General Surgery, The Second Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
28
|
Park W, Cho S, Han J, Shin H, Na K, Lee B, Kim DH. Advanced smart-photosensitizers for more effective cancer treatment. Biomater Sci 2017; 6:79-90. [PMID: 29142997 PMCID: PMC5736440 DOI: 10.1039/c7bm00872d] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Photodynamic therapy (PDT) based upon the use of light and photosensitizers (PSs) has been used as a novel treatment approach for a variety of tumors. It, however, has several major limitations in the clinic: poor water solubility, long-term phototoxicity, low tumor targeting efficacy, and limited light penetration. With advances in nanotechnology, materials science, and clinical interventional imaging procedures, various smart-PSs have been developed for improving their cancer-therapeutic efficacy while reducing the adverse effects. Here, we briefly review state-of-the-art smart-PSs and discuss the future directions of PDT technology.
Collapse
Affiliation(s)
- Wooram Park
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Soojeong Cho
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Jieun Han
- Center for Photomedicine, Department of Biotechnology, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Heejun Shin
- Center for Photomedicine, Department of Biotechnology, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kun Na
- Center for Photomedicine, Department of Biotechnology, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Byeongdu Lee
- X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
29
|
Schmohl KA, Gupta A, Grünwald GK, Trajkovic-Arsic M, Klutz K, Braren R, Schwaiger M, Nelson PJ, Ogris M, Wagner E, Siveke JT, Spitzweg C. Imaging and targeted therapy of pancreatic ductal adenocarcinoma using the theranostic sodium iodide symporter (NIS) gene. Oncotarget 2017; 8:33393-33404. [PMID: 28380420 PMCID: PMC5464876 DOI: 10.18632/oncotarget.16499] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/27/2017] [Indexed: 12/18/2022] Open
Abstract
The theranostic sodium iodide symporter (NIS) gene allows detailed molecular imaging of transgene expression and application of therapeutic radionuclides. As a crucial step towards clinical application, we investigated tumor specificity and transfection efficiency of epidermal growth factor receptor (EGFR)-targeted polyplexes as systemic NIS gene delivery vehicles in an advanced genetically engineered mouse model of pancreatic ductal adenocarcinoma (PDAC) that closely reflects human disease. PDAC was induced in mice by pancreas-specific activation of constitutively active KrasG12D and deletion of Trp53. We used tumor-targeted polyplexes (LPEI-PEG-GE11/NIS) based on linear polyethylenimine, shielded by polyethylene glycol and coupled with the EGFR-specific peptide ligand GE11, to target a NIS-expressing plasmid to high EGFR-expressing PDAC. In vitro iodide uptake studies in cell explants from murine EGFR-positive and EGFR-ablated PDAC lesions demonstrated high transfection efficiency and EGFR-specificity of LPEI-PEG-GE11/NIS. In vivo 123I gamma camera imaging and three-dimensional high-resolution 124I PET showed significant tumor-specific accumulation of radioiodide after systemic LPEI-PEG-GE11/NIS injection. Administration of 131I in LPEI-PEG-GE11/NIS-treated mice resulted in significantly reduced tumor growth compared to controls as determined by magnetic resonance imaging, though survival was not significantly prolonged. This study opens the exciting prospect of NIS-mediated radionuclide imaging and therapy of PDAC after systemic non-viral NIS gene delivery.
Collapse
Affiliation(s)
- Kathrin A. Schmohl
- Department of Internal Medicine II and IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Aayush Gupta
- Department of Internal Medicine II, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Geoffrey K. Grünwald
- Department of Internal Medicine II and IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Marija Trajkovic-Arsic
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin Klutz
- Department of Internal Medicine II and IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Rickmer Braren
- Department of Radiology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Peter J. Nelson
- Clinical Biochemistry Group, Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Manfred Ogris
- Department of Pharmaceutical Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for System-Based Drug Research and Center for Nanoscience, LMU Munich, Munich, Germany
| | - Jens T. Siveke
- Department of Internal Medicine II, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Spitzweg
- Department of Internal Medicine II and IV, University Hospital of Munich, LMU Munich, Munich, Germany
| |
Collapse
|
30
|
Cheng P, Zeng W, Li L, Huo D, Zeng L, Tan J, Zhou J, Sun J, Liu G, Li Y, Guan G, Wang Y, Zhu C. PLGA-PNIPAM Microspheres Loaded with the Gastrointestinal Nutrient NaB Ameliorate Cardiac Dysfunction by Activating Sirt3 in Acute Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2016; 3:1600254. [PMID: 27981013 PMCID: PMC5157182 DOI: 10.1002/advs.201600254] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/22/2016] [Indexed: 05/06/2023]
Abstract
Acute myocardial infarction (AMI) is the death of cardiomyocytes caused by a lack of energy due to ischemia. Nutrients supplied by the blood are the main source of cellular energy for cardiomyocytes. Sodium butyrate (NaB), a gastrointestinal nutrient, is a short-chain fatty acid (butyric acid) that may act as an energy source in AMI therapy. Poly(lactic-co-glycolic acid)-Poly (N-isopropylacrylamide) microspheres loaded with NaB (PP-N) are synthesized to prolong the release of NaB and are injected into ischemic zones in a Sprague-Dawley rat AMI model. Here, this study shows that PP-N can significantly ameliorate cardiac dysfunction in AMI, and NaB can specially bind to Sirt3 structure, activating its deacetylation ability and inhibiting the generation of reactive oxygen species, autophagy, and angiogenesis promotion. The results indicate that NaB, acting as a nutrient, can protect cardiomyocytes in AMI. These results suggest that the gastrointestinal nutrient NaB may be a new therapy for AMI treatment, and PP-N may be the ideal therapeutic regimen.
Collapse
Affiliation(s)
- Panke Cheng
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Wen Zeng
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Li Li
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Da Huo
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Lingqing Zeng
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Ju Tan
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Jingting Zhou
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Jiansen Sun
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Ge Liu
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Yanzhao Li
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Ge Guan
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Yuxin Wang
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| | - Chuhong Zhu
- Department of AnatomyNational & Regional Engineering Laboratory of Tissue EngineeringState and Local Joint Engineering Laboratory For Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of ChongqingThird Military Medical UniversityChongqing400038China
| |
Collapse
|
31
|
Wang J, Cui H. Nanostructure-Based Theranostic Systems. Theranostics 2016; 6:1274-6. [PMID: 27375778 PMCID: PMC4924498 DOI: 10.7150/thno.16479] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022] Open
Abstract
This themed issue continues the focus on the recent developments of nanoscaled theranostic systems for early and accurate disease diagnosis, especially cancer diagnosis, as well as effective cancer treatment and management.
Collapse
Affiliation(s)
- Jun Wang
- 1. Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei Anhui 230027, PR China
- 2. CAS Center for Excellence in Nanoscience, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei Anhui 230027, PR China
- 3. Innovation Center for Cell Signaling Network, University of Science and Technology of China, Hefei, Anhui 230027, PR China
- ✉ Corresponding authors: ;
| | - Honggang Cui
- 4. Department of Chemical and Biomolecular Engineering, and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- 5. Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- 6. Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
- ✉ Corresponding authors: ;
| |
Collapse
|