1
|
Liu X, Shao Y, Li Y, Chen Z, Shi T, Tong Q, Zou X, Ju L, Pan J, Zhuang R, Pan X. Extensive Review of Nanomedicine Strategies Targeting the Tumor Microenvironment in PDAC. Int J Nanomedicine 2025; 20:3379-3406. [PMID: 40125427 PMCID: PMC11927507 DOI: 10.2147/ijn.s504503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world, mainly because of its powerful pro-connective tissue proliferation matrix and immunosuppressive tumor microenvironment (TME), which promote tumor progression and metastasis. In addition, the extracellular matrix leads to vascular collapse, increased interstitial fluid pressure, and obstruction of lymphatic return, thereby hindering effective drug delivery, deep penetration, and immune cell infiltration. Therefore, reshaping the TME to enhance tumor perfusion, increase deep drug penetration, and reverse immune suppression has become a key therapeutic strategy. Traditional therapies for PDAC, including surgery, radiation, and chemotherapy, face significant limitations. Surgery is challenging due to tumor location and growth, while chemotherapy and radiation are hindered by the dense extracellular matrix and immunosuppressive TME. In recent years, the advancement of nanotechnology has provided new opportunities to improve drug efficacy. Nanoscale drug delivery systems (NDDSs) provide several advantages, including improved drug stability in vivo, enhanced tumor penetration, and reduced systemic toxicity. However, the clinical translation of nanotechnology in PDAC therapy faces several challenges. These include the need for precise targeting and control over drug release, potential immune responses to the nanocarriers, and the scalability and cost-effectiveness of production. This article provides an overview of the latest nanobased methods for achieving better therapeutic outcomes and overcoming drug resistance. We pay special attention to TME-targeted therapy in the context of PDAC, discuss the advantages and limitations of current strategies, and emphasize promising new developments. By emphasizing the enormous potential of NDDSs in improving the treatment outcomes of patients with PDAC, while critically discussing the limitations of traditional therapies and the challenges faced by nanotechnology in achieving clinical breakthroughs, our review paves the way for future research in this rapidly developing field.
Collapse
Affiliation(s)
- Xing Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, People’s Republic of China
| | - Yidan Shao
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Yunjiang Li
- Radiology Department, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Zuhua Chen
- Radiology Department, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Tingting Shi
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Qiao Tong
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Xi Zou
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Liping Ju
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Jinming Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Xuwang Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| |
Collapse
|
2
|
Lu W, Li Y, Zhang X, Wang N, Chen D, Zhao Y, Li G, Shi X, Ma X, Su X, Wang F, Shu C, Chen K. Dual-modal overcoming of physical barriers for improved photodynamic cancer therapy via soft organosilica nanocapsules. J Nanobiotechnology 2024; 22:734. [PMID: 39593093 PMCID: PMC11600580 DOI: 10.1186/s12951-024-02945-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024] Open
Abstract
Amidst the burgeoning field of cancer nanomedicine, dense extracellular matrices and anomalous vascular structures in the tumor microenvironment (TME) present substantial physical barriers to effective therapeutic delivery. These physical barriers hinder the optimal bioavailability of nanomedicine. Here, we propose a pioneering dual-modal strategy for overcoming physical barriers via soft organosilica nanocapsules (SMONs). Hyaluronidase-modified flexible spheres work by degrading the extracellular matrix and utilizing their flexible characteristics to enhance penetration into deeper layers. Compared with their stiff counterparts, the SMONs show diminished Young's modulus, then the inherent softness of the SMONs confers distinct advantages, and significantly augmented cellular internalization within 4T1 cells, leading to an amplified in vitro photodynamic therapeutic effect. Furthermore, hyaluronidase-functionalized SMONs (SMONs-HAase) exhibit enhanced tumor penetration in 3D spheroids. Post incorporation of the photosensitizer chlorin e6, when administered intravenously, these soft organosilica nanocapsules amplify the efficacy of photodynamic therapy. In addition, RNA-seq analysis of SMONs-HAase-Ce6 shows it alters gene expression, degrading the extracellular matrix and impairing mitochondrial function. To sum up, this work elucidates the potential of a dual-modal strategy, highlighting the promise of SMONs in overcoming TME physical barriers and optimizing therapeutic outcomes.
Collapse
Affiliation(s)
- Wei Lu
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, P.R. China
| | - Xiaojun Zhang
- Department of Radiology, Children's Hospital of Nanjing Medical University, Nanjing, 210018, P.R. China
| | - Ning Wang
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Dong Chen
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Yatong Zhao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 213161, P.R. China
| | - Guang Li
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410000, P.R. China
| | - Xuzhi Shi
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Xiaobo Ma
- College of Electronic and Optical Engineering & College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China
| | - Feng Wang
- Department of Radiology, Children's Hospital of Nanjing Medical University, Nanjing, 210018, P.R. China.
| | - Chuqiang Shu
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410000, P.R. China.
| | - Kun Chen
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P.R. China.
| |
Collapse
|
3
|
Wang Y, Zhou Q, Luo W, Yang X, Zhang J, Lou Y, Mao J, Chen J, Wu F, Hou J, Tang G, Bai H, Yu R. A collagenase-decorated Cu-based nanotheranostics: remodeling extracellular matrix for optimizing cuproptosis and MRI in pancreatic ductal adenocarcinoma. J Nanobiotechnology 2024; 22:689. [PMID: 39523309 PMCID: PMC11552245 DOI: 10.1186/s12951-024-02968-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), characterized by a dense extracellular matrix (ECM), presents significant therapeutic challenges due to its poor prognosis and high resistance to chemotherapy. Current chemodrugs and diagnostic agents largely fail to cross the barrier posed by the ECM, which severely limits the PDAC theranostics. This study introduces a novel theranostic strategy using thioether-hybridized hollow mesoporous organosilica nanoparticles (dsMNs) for the co-delivery of copper (Cu) and disulfiram (DSF), aiming to induce cuproptosis in PDAC cells. Our approach leverages the ECM-degrading enzyme collagenase, integrated with dsMNs, to enhance drug penetration by reducing matrix stiffness. Furthermore, the innovative use of a pancreatic cancer cell membrane coating on the nanoparticles enhances tumor targeting and stability (dsMCu-D@M-Co). The multifunctional platform not only facilitates deep drug penetration and triggers cuproptosis effectively but also utilizes the inherent properties of Cu to serve as a T1-weighted magnetic resonance imaging (MRI) contrast agent. In vitro and in vivo assessments demonstrate significant tumor size reduction in PDAC-bearing mice, highlighting the dual functionality of our platform in improving therapeutic efficacy and diagnostic precision. This integrated strategy represents a significant advancement in the management of PDAC, offering a promising new direction for overcoming one of the most lethal cancers.
Collapse
Affiliation(s)
- Yining Wang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Qiaomei Zhou
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Wangping Luo
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Xiaoyan Yang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Jinguo Zhang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Yijie Lou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Jin Mao
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Jiayi Chen
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China
| | - Fan Wu
- Department of Neurosurgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Jue Hou
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China
| | - Guping Tang
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China
| | - Hongzhen Bai
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China.
| | - Risheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China.
| |
Collapse
|
4
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [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: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
5
|
He Y, Feng Y, Qiu D, Lin M, Jin H, Hu Z, Huang X, Ma S, He Y, Lai M, Jin W, Liu J. Regulation of IFP in solid tumours through acoustic pressure to enhance infiltration of nanoparticles of various sizes. J Drug Target 2024; 32:964-976. [PMID: 38884143 DOI: 10.1080/1061186x.2024.2367579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Numerous nanomedicines have been developed recently that can accumulate selectively in tumours due to the enhanced permeability and retention (EPR) effect. However, the high interstitial fluid pressure (IFP) in solid tumours limits the targeted delivery of nanomedicines. We were previously able to relieve intra-tumoural IFP by low-frequency non-focused ultrasound (LFNFU) through ultrasonic targeted microbubble destruction (UTMD), improving the targeted delivery of FITC-dextran. However, the accumulation of nanoparticles of different sizes and the optimal acoustic pressure were not evaluated. In this study, we synthesised Cy5.5-conjugated mesoporous silica nanoparticles (Cy5.5-MSNs) of different sizes using a one-pot method. The Cy5.5-MSNs exhibited excellent stability and biosafety regardless of size. MCF7 tumour-bearing mice were subjected to UTMD over a range of acoustic pressures (0.5, 0.8, 1.5 and 2.0 MPa), and injected intravenously with Cy5.5-MSNs. Blood perfusion, tumour IFP and intra-tumoural accumulation of Cy5.5-MSNs were analysed. Blood perfusion and IFP initially rose, and then declined, as acoustic pressure intensified. Furthermore, UTMD significantly enhanced the accumulation of differentially sized Cy5.5-MSNs in tumour tissues compared to that of the control group, and the increase was sevenfold higher at an acoustic pressure of 1.5 MPa. Taken together, UTMD enhanced the infiltration and accumulation of Cy5.5-MSNs of different sizes in solid tumours by reducing intra-tumour IFP.
Collapse
Affiliation(s)
- Yangcheng He
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Yuyi Feng
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Danxai Qiu
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - MinHua Lin
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Hai Jin
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Zhiwen Hu
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Xue Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Suihong Ma
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Yan He
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Meiqi Lai
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Wenhui Jin
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Jianhua Liu
- Department of Ultrasound, School of Medicine, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
6
|
Zhou Y, Ma Y, Sheng J, Ma Y, Ding J, Zhou W. Breaking Down Barriers in Drug Delivery by Stromal Remodeling Approaches in Pancreatic Cancer. Mol Pharm 2024; 21:3764-3776. [PMID: 39049481 DOI: 10.1021/acs.molpharmaceut.4c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Pancreatic cancer remains a formidable challenge in oncology due to its aggressive nature and limited treatment options. The dense stroma surrounding pancreatic tumors not only provides structural support but also presents a formidable barrier to effective therapy, hindering drug penetration and immune cell infiltration. This review delves into the intricate interplay between stromal components and cancer cells, highlighting their impact on treatment resistance and prognosis. Strategies for stromal remodeling, including modulation of cancer-associated fibroblasts (CAFs), pancreatic stellate cells (PSCs) activation states, and targeting extracellular matrix (ECM) components, are examined for their potential to enhance drug penetration and improve therapeutic efficacy. Integration of stromal remodeling with conventional therapies, such as chemotherapy and immunotherapy, is discussed along with the emerging field of intelligent nanosystems for targeted drug delivery. This comprehensive overview underscores the importance of stromal remodeling in pancreatic cancer treatment and offers insights into promising avenues for future research and clinical translation.
Collapse
Affiliation(s)
- Ying Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yunxiao Ma
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Jianwei Sheng
- China Quality Mark Certification (Shandong) Co., LTD, Jinan, Shandong 250100, China
| | - Yiran Ma
- Hunan Bainianyiren Chinese Traditional Medical Institute Co., LTD, Changsha, Hunan 410221, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha, Hunan 410008, China
| |
Collapse
|
7
|
Ferdous KU, Tesfay MZ, Cios A, Shelton RS, Hartupee C, Urbaniak A, Chamcheu JC, Mavros MN, Giorgakis E, Mustafa B, Simoes CC, Miousse IR, Basnakian AG, Moaven O, Post SR, Cannon MJ, Kelly T, Nagalo BM. Enhancing Neoadjuvant Virotherapy's Effectiveness by Targeting Stroma to Improve Resectability in Pancreatic Cancer. Biomedicines 2024; 12:1596. [PMID: 39062169 PMCID: PMC11275208 DOI: 10.3390/biomedicines12071596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
About one-fourth of patients with pancreatic ductal adenocarcinoma (PDAC) are categorized as borderline resectable (BR) or locally advanced (LA). Chemotherapy and radiation therapy have not yielded the anticipated outcomes in curing patients with BR/LA PDAC. The surgical resection of these tumors presents challenges owing to the unpredictability of the resection margin, involvement of vasculature with the tumor, the likelihood of occult metastasis, a higher ratio of positive lymph nodes, and the relatively larger size of tumor nodules. Oncolytic virotherapy has shown promising activity in preclinical PDAC models. Unfortunately, the desmoplastic stroma within the PDAC tumor microenvironment establishes a barrier, hindering the infiltration of oncolytic viruses and various therapeutic drugs-such as antibodies, adoptive cell therapy agents, and chemotherapeutic agents-in reaching the tumor site. Recently, a growing emphasis has been placed on targeting major acellular components of tumor stroma, such as hyaluronic acid and collagen, to enhance drug penetration. Oncolytic viruses can be engineered to express proteolytic enzymes that cleave hyaluronic acid and collagen into smaller polypeptides, thereby softening the desmoplastic stroma, ultimately leading to increased viral distribution along with increased oncolysis and subsequent tumor size regression. This approach may offer new possibilities to improve the resectability of patients diagnosed with BR and LA PDAC.
Collapse
Affiliation(s)
- Khandoker Usran Ferdous
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Mulu Z. Tesfay
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Aleksandra Cios
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
| | - Randal S. Shelton
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Conner Hartupee
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA; (C.H.); (O.M.)
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.U.); (I.R.M.)
| | - Jean Christopher Chamcheu
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA;
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michail N. Mavros
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Emmanouil Giorgakis
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Bahaa Mustafa
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Camila C. Simoes
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Isabelle R. Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.U.); (I.R.M.)
| | - Alexei G. Basnakian
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Central Arkansas Veterans Healthcare System, John L. McClellan Memorial VA Hospital, Little Rock, AR 72205, USA
| | - Omeed Moaven
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA; (C.H.); (O.M.)
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA 70112, USA
| | - Steven R. Post
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Martin J. Cannon
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Thomas Kelly
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.U.F.); (M.Z.T.); (A.C.); (C.C.S.); (S.R.P.); (T.K.)
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.N.M.); (M.J.C.)
| |
Collapse
|
8
|
Hong L, Xu K, Yang M, Zhu L, Chen C, Xu L, Zhu W, Jin L, Wang L, Lin J, Wang J, Ren W, Wu A. VISTA antibody-loaded Fe 3O 4@TiO 2 nanoparticles for sonodynamic therapy-synergistic immune checkpoint therapy of pancreatic cancer. Mater Today Bio 2024; 26:101106. [PMID: 38883421 PMCID: PMC11176928 DOI: 10.1016/j.mtbio.2024.101106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Breaking the poor permeability of immune checkpoint inhibitors (ICIs) caused by the stromal barrier and reversing the immunosuppressive microenvironment are significant challenges in pancreatic cancer immunotherapy. In this study, we synthesized core-shell Fe3O4@TiO2 nanoparticles to act as carriers for loading VISTA monoclonal antibodies to form Fe3O4@TiO2@VISTAmAb (FTV). The nanoparticles are designed to target the overexpressed ICIs VISTA in pancreatic cancer, aiming to improve magnetic resonance imaging-guided sonodynamic therapy (SDT)-facilitated immunotherapy. Laser confocal microscopy and flow cytometry results demonstrate that FTV nanoparticles are specifically recognized and phagocytosed by Panc-2 cells. In vivo experiments reveal that ultrasound-triggered TiO2 SDT can induce tumor immunogenic cell death (ICD) and recruit T-cell infiltration within the tumor microenvironment by releasing damage-associated molecular patterns (DAMPs). Furthermore, ultrasound loosens the dense fibrous stroma surrounding the pancreatic tumor and increases vascular density, facilitating immune therapeutic efficiency. In summary, our study demonstrates that FTV nanoparticles hold great promise for synergistic SDT and immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Lu Hong
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Kaiwei Xu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Ming Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Lubing Zhu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Chunqu Chen
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Liu Xu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
| | - Weihao Zhu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Lufei Jin
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
| | - Linwei Wang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Jie Lin
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, PR China
| | - Jianhua Wang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Wenzhi Ren
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, PR China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, PR China
| |
Collapse
|
9
|
Nene LC, Abrahamse H. Design consideration of phthalocyanines as sensitizers for enhanced sono-photodynamic combinatorial therapy of cancer. Acta Pharm Sin B 2024; 14:1077-1097. [PMID: 38486981 PMCID: PMC10935510 DOI: 10.1016/j.apsb.2023.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/08/2023] [Accepted: 11/25/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer remains one of the diseases with the highest incidence and mortality globally. Conventional treatment modalities have demonstrated threatening drawbacks including invasiveness, non-controllability, and development of resistance for some, including chemotherapy, radiation, and surgery. Sono-photodynamic combinatorial therapy (SPDT) has been developed as an alternative treatment modality which offers a non-invasive and controllable therapeutic approach. SPDT combines the mechanism of action of sonodynamic therapy (SDT), which uses ultrasound, and photodynamic therapy (PDT), which uses light, to activate a sensitizer and initiate cancer eradication. The use of phthalocyanines (Pcs) as sensitizers for SPDT is gaining interest owing to their ability to induce intracellular oxidative stress and initiate toxicity under SDT and PDT. This review discusses some of the structural prerequisites of Pcs which may influence their overall SPDT activities in cancer therapy.
Collapse
Affiliation(s)
- Lindokuhle Cindy Nene
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein 2028, South Africa
| |
Collapse
|
10
|
Xiang Y, Chen Q, Nan Y, Liu M, Xiao Z, Yang Y, Zhang J, Ying X, Long X, Wang S, Sun J, Huang Q, Ai K. Nitric Oxide‐Based Nanomedicines for Conquering TME Fortress: Say “NO” to Insufficient Tumor Treatment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202312092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Indexed: 01/02/2025]
Abstract
AbstractAlmost all cancer treatments are significantly limited by the strong tumor microenvironment (TME) fortress formed by abnormal vasculature, dense extracellular matrix (ECM), multidrug resistance (MDR) system, and immune “cold” environment. In the huge efforts of dismantling the TME fortress, nitric oxide (NO)‐based nanomedicines are increasingly occupying a central position and have already been identified as super “strong polygonal warriors” to dismantle TME fortress for efficient cancer treatment, benefiting from NO's unique physicochemical properties and extremely fascinating biological effects. However, there is a paucity of systematic review to elaborate on the progress and fundamental mechanism of NO‐based nanomedicines in oncology from this aspect. Herein, the key characteristics of TME fortress and the potential of NO in reprogramming TME are delineated and highlighted. The evolution of NO donors and the advantages of NO‐based nanomedicines are discussed subsequently. Moreover, the latest progress of NO‐based nanomedicines for solid tumors is comprehensively reviewed, including normalizing tumor vasculature, overcoming ECM barrier, reversing MDR, and reactivating the immunosuppression TME. Lastly, the prospects, limitations, and future directions on NO‐based nanomedicines for TME manipulation are discussed to provide new insights into the construction of more applicable anticancer nanomedicines.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yayun Nan
- Geriatric Medical Center People's Hospital of Ningxia Hui Autonomous Region Yinchuan Ningxia 750002 P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yuqi Yang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Jinping Zhang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Xingyu Long
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Jian Sun
- College of Pharmacy Xinjiang Medical University Urumqi 830017 P. R. China
| | - Qiong Huang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and Treatment Ministry of Education Xiangya Hospital Central South University Changsha 410078 P. R. China
| |
Collapse
|
11
|
Zlotver I, Sosnik A. Glucosylated Hybrid TiO 2 /Polymer Nanomaterials for Actively Targeted Sonodynamic Therapy of Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305475. [PMID: 37715267 DOI: 10.1002/smll.202305475] [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: 06/30/2023] [Revised: 08/29/2023] [Indexed: 09/17/2023]
Abstract
Sonodynamic therapy (SDT) is an anti-cancer therapeutic strategy based on the generation of reactive oxygen species (ROS) upon local ultrasound (US) irradiation of sono-responsive molecules or nanomaterials that accumulate in the tumor. In this work, the sonodynamic efficiency of sono-responsive hybrid nanomaterials composed of amorphous titanium dioxide and an amphiphilic poly(ethylene oxide)-b-poly(propylene oxide) block copolymer is synthesized, fully characterized, and investigated both in vitro and in vivo. The modular and versatile synthetic pathway enables the control of the nanoparticle size between 30 and 300 nm (dynamic light scattering) and glucosylation of the surface for active targeting of tumors overexpressing glucose transporters. Studies on 2D and 3D rhabdomyosarcoma cell cultures reveal a statistically significant increase in the sonodynamic efficiency of glucosylated hybrid nanoparticles with respect to unmodified ones. Using a xenograft rhabdomyosarcoma murine model, it is demonstrated that by tuning the nanoparticle size and surface features, the tumor accumulation is increased by ten times compared to main off-target clearance organs such as the liver. Finally, the SDT of rhabdomyosarcoma-bearing mice is investigated with 50-nm glucosylated nanoparticles. Findings evidence a dramatic prolongation of the animal survival and tumor volumes 100 times smaller than those treated only with ultrasound or nanoparticles.
Collapse
Affiliation(s)
- Ivan Zlotver
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
12
|
Cheng P, Ming S, Cao W, Wu J, Tian Q, Zhu J, Wei W. Recent advances in sonodynamic therapy strategies for pancreatic cancer. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1945. [PMID: 38403882 DOI: 10.1002/wnan.1945] [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: 09/25/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Pancreatic cancer, a prevalent malignancy of the digestive system, has a poor 5-year survival rate of around 10%. Although numerous minimally invasive alternative treatments, including photothermal therapy and photodynamic therapy, have shown effectiveness compared with traditional surgical procedures, radiotherapy, and chemotherapy. However, the application of these alternative treatments is constrained by their depth of penetration, making it challenging to treat pancreatic cancer situated deep within the tissue. Sonodynamic therapy (SDT) has emerged as a promising minimally invasive therapy method that is particularly potent against deep-seated tumors such as pancreatic cancer. However, the unique characteristics of pancreatic cancer, including a dense surrounding matrix, high reductivity, and a hypoxic tumor microenvironment, impede the efficient application of SDT. Thus, to guide the evolution of SDT for pancreatic cancer therapy, this review addresses these challenges, examines current strategies for effective SDT enhancement for pancreatic cancer, and investigates potential future advances to boost clinical applicability. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuai Ming
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Cao
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jixiao Wu
- School of Materials and Chemistry, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jing Zhu
- School of Materials and Chemistry, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Wei Wei
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
13
|
Zhang Z, Yuan Y, Xue Y, Zhang W, Sun X, Xu X, Liu C. Nanomaterials for Ultrasound Imaging- Guided Sonodynamic Therapy. Technol Cancer Res Treat 2024; 23:15330338241263197. [PMID: 39051705 PMCID: PMC11273702 DOI: 10.1177/15330338241263197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 07/27/2024] Open
Abstract
Ultrasound examination is becoming the most popular medical imaging modality because of its low cost and high safety profile. Ultrasound contrast agents enhance the scattering of sound waves, which can improve the clarity and resolution of images. Nanoparticle Ultrasound contrast agents have the characteristics of a large specific surface area and a modifiable surface, which can increase drug loading capacity, prolong circulation time, and enable drug enrichment in specific organs or tissues. This leads to improved therapeutic effects and reducing toxic and side effects. Compared with traditional ultrasound contrast agents, Nano-ultrasound contrast agents overcome the limitation of imaging solely within blood vessels and facilitate imaging within tumor tissues, thereby extending the duration of enhanced imaging. Sonodynamic therapy is an emerging treatment method that has been developed rapidly in recent years, which has the advantages of noninvasive, high spatial and temporal resolution, and low toxicity and side effects. Sonodynamic therapy utilizes a sonosensitizer that, when excited by ultrasound at the tumor site, produces toxic reactive oxygen species, inducing apoptosis or necrosis in tumor cells. Ultrasound-guided sonodynamic therapy allows for real-time observation of lesions, is convenient and flexible, and is free of radiation exposure. With the use of nanomaterials as carriers, ultrasound-guided sonodynamic therapy has made significant strides. This study categorizes and summarizes the current research on acoustic sensitizer carrier materials, including carbon-based, silicon-based, peptide-based, iron-based, metal-organic frameworks, polymers, and liposomes. It concludes by highlighting the current challenges in the integration of ultrasound imaging with sonodynamic therapy and suggests future directions for clinical application development.
Collapse
Affiliation(s)
- Zhiyang Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yinuo Yuan
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanzhang Xue
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenjing Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao Sun
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, China
| | - Cun Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
14
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
15
|
Zhou R, Chang M, Shen M, Cong Y, Chen Y, Wang Y. Sonocatalytic Optimization of Titanium-Based Therapeutic Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301764. [PMID: 37395421 PMCID: PMC10477905 DOI: 10.1002/advs.202301764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/28/2023] [Indexed: 07/04/2023]
Abstract
Recent considerable technological advances in ultrasound-based treatment modality provides a magnificent prospect for scientific communities to conquer the related diseases, which is featured with remarkable tissue penetration, non-invasive and non-thermal characteristics. As one of the critical elements that influences treatment outcomes, titanium (Ti)-based sonosensitizers with distinct physicochemical properties and exceptional sonodynamic efficiency have been applied extensively in the field of nanomedical applications. To date, a myriad of methodologies has been designed to manipulate the sonodynamic performance of titanium-involved nanomedicine and further enhance the productivity of reactive oxygen species for disease treatments. In this comprehensive review, the sonocatalytic optimization of diversified Ti-based nanoplatforms, including defect engineering, plasmon resonance modulation, heterojunction, modulating tumor microenvironment, as well as the development of synergistic therapeutic modalities is mainly focused. The state-of-the-art Ti-based nanoplatforms ranging from preparation process to the extensive medical applications are summarized and highlighted, with the goal of elaborating on future research prospects and providing a perspective on the bench-to-beside translation of these sonocatalytic optimization tactics. Furthermore, to spur further technological advancements in nanomedicine, the difficulties currently faced and the direction of sonocatalytic optimization of Ti-based therapeutic nanomedicine are proposed and outlooked.
Collapse
Affiliation(s)
- Ruirui Zhou
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| | - Meiqi Chang
- Laboratory CenterShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071P. R. China
| | - Mengjun Shen
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| | - Yang Cong
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yin Wang
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| |
Collapse
|
16
|
Zhang H, Wang Y, Zhu L, Qi Z, Cao K, Chang J, Hou L. A "bulldozer" driven by anoxic bacteria for pancreatic cancer chemo-immunotherapy. J Control Release 2023; 360:660-671. [PMID: 37433371 DOI: 10.1016/j.jconrel.2023.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023]
Abstract
Immune evasion is a major obstacle for pancreatic ductal adenocarcinoma (PDAC) therapy. Inhibition of autophagy can improve antigen presentation and enlarge immunogenic cell death (ICD) effect to generate a strong anti-tumor immune response. However, abundant extracellular matrix dominated by hyaluronic acid (HA) hinders the deep penetration of autophagy inhibitors and ICD inducers. Herein, an intelligent autophagy inhibitor hydroxychloroquine (HCQ) and chemotherapeutic drug doxorubicin (DOX) co-loaded "bulldozer" (HD@HH/EcN) driven by anoxic bacteria was constructed for PDAC chemo-immunotherapy. Results demonstrated that probiotic Escherichia coli 1917 (EcN) could carry hyaluronidases (HAases)-hybrided albumin nanoparticles (HD@HH) to reach PDAC tumor tissue quickly and accurately. Thereafter, HAases can efficiently cleave the tumor matrix barrier and promote HD@HH/EcN to accumulate at tumor hypoxic core significantly. After that, high level of glutathione (GSH) in tumor microenvironment (TME) induces intermolecular disulfide bond in HD@HH nanoparticles breakage, to precisely release HCQ and DOX. DOX can induce ICD effect. Meanwhile, HCQ can amplify DOX induced ICD effect by inhibiting tumor autophagy, which further increase cell surface expression of major histocompatibility complex class I (MHC-I) and augment recruitment of CD8+ T cell to improve immunosuppressive TME. This study provides a new strategy for PDAC chemo-immunotherapy.
Collapse
Affiliation(s)
- Huijuan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China.
| | - Yaping Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ling Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China
| | - Zijun Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Kexuan Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junbiao Chang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China; School of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China.
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China.
| |
Collapse
|
17
|
He M, Yu H, Zhao Y, Liu J, Dong Q, Xu Z, Kang Y, Xue P. Ultrasound-Activatable g-C 3 N 4 -Anchored Titania Heterojunction as an Intracellular Redox Homeostasis Perturbator for Augmented Oncotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300244. [PMID: 36843276 DOI: 10.1002/smll.202300244] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/11/2023] [Indexed: 05/25/2023]
Abstract
Energy band structure of inorganic nano-sonosensitizers is usually optimized by surface decoration with noble metals or metal oxide semiconductors, aiming to enhance interfacial charge transfer, augment spin-flip and promote radical generation. To avoid potential biohazards of metallic elements, herein, metal-free graphitic carbon nitride quantum dots (g-C3 N4 QDs) are anchored onto hollow mesoporous TiO2 nanostructure to formulate TiO2 @g-C3 N4 heterojunction. The direct Z-scheme charge transfer significantly improves the separation/recombination dynamics of electron/hole (e- /h+ ) pairs upon ultrasound (US) stimulation, which promotes the yield of singlet oxygen (1 O2 ) and hydroxyl radicals (·OH). The conjugated g-C3 N4 QDs with peroxidase-mimic activity further react with the elevated endogenous H2 O2 and aggravate oxidative stress. After loading prodrug romidepsin (RMD) in TiO2 @g-C3 N4 , stimulus-responsive drug delivery can be realized by US irradiation. The disulfide bridge of the released RMD tends to be reduced by glutathione (GSH) into a monocyclic dithiol, which arrests cell cycle in G2/M phase and evokes apoptosis through enhanced histone acetylation. Importantly, reactive oxygen species accumulation accompanied by GSH depletion is devoted to deleterious redox dyshomeostasis, leading to augmented systemic oncotherapy by eliciting antitumor immunity. Collectively, this paradigm provides useful insights in optimizing the performance of TiO2 -based nano-sonosensitizers for tackling critical diseases.
Collapse
Affiliation(s)
- Mengting He
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Honglian Yu
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Yinmin Zhao
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Jiahui Liu
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Qi Dong
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Zhigang Xu
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Yuejun Kang
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| | - Peng Xue
- School of Materials and Energy, Southwest University, Chongqing, 400715, China
| |
Collapse
|
18
|
Naletova I, Tomasello B, Attanasio F, Pleshkan VV. Prospects for the Use of Metal-Based Nanoparticles as Adjuvants for Local Cancer Immunotherapy. Pharmaceutics 2023; 15:1346. [PMID: 37242588 PMCID: PMC10222518 DOI: 10.3390/pharmaceutics15051346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Immunotherapy is among the most effective approaches for treating cancer. One of the key aspects for successful immunotherapy is to achieve a strong and stable antitumor immune response. Modern immune checkpoint therapy demonstrates that cancer can be defeated. However, it also points out the weaknesses of immunotherapy, as not all tumors respond to therapy and the co-administration of different immunomodulators may be severely limited due to their systemic toxicity. Nevertheless, there is an established way through which to increase the immunogenicity of immunotherapy-by the use of adjuvants. These enhance the immune response without inducing such severe adverse effects. One of the most well-known and studied adjuvant strategies to improve immunotherapy efficacy is the use of metal-based compounds, in more modern implementation-metal-based nanoparticles (MNPs), which are exogenous agents that act as danger signals. Adding innate immune activation to the main action of an immunomodulator makes it capable of eliciting a robust anti-cancer immune response. The use of an adjuvant has the peculiarity of a local administration of the drug, which positively affects its safety. In this review, we will consider the use of MNPs as low-toxicity adjuvants for cancer immunotherapy, which could provide an abscopal effect when administered locally.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, 95125 Catania, Italy
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| |
Collapse
|
19
|
Yang M, Ren W, Cui H, Qin Q, Wang Q, Zhu W, Wu X, Pan C, Qi X, Wu A. Ginsenoside Rk1-Loaded Manganese-Doped Hollow Titania for Enhancing Tumor Sonodynamic Therapy via Upregulation of Intracellular Reactive Oxygen Species. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20800-20810. [PMID: 37078779 DOI: 10.1021/acsami.3c03476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Amplifying the intracellular reactive oxygen species (ROS) level remains an urgent challenge for efficient sonodynamic therapy (SDT) of tumors. Herein, by loading ginsenoside Rk1 with manganese-doped hollow titania (MHT), a Rk1@MHT sonosensitizer was conceived to strengthen the outcome of tumor SDT. The results verify that manganese-doping remarkably elevates the UV-visible absorption and decreases the bandgap energy of titania from 3.2 to 3.0 eV, which improves ROS production under ultrasonic irradiation. Immunofluorescence and Western blot analysis demonstrate that ginsenoside Rk1 can block the critical protein of the glutathione synthesis pathway, glutaminase, thus enhancing intracellular ROS by eliminating the endogenous glutathione-depleted pathway of ROS. Manganese-doping confers the nanoprobe T1-weighted MRI function (r2/r1 = 1.41). Moreover, the in vivo tests confirm that Rk1@MHT-based SDT eradicates liver cancer in tumor-bearing mice via dual upregulation of intracellular ROS production. In summary, our study provides a new strategy for designing high-performance sonosensitizer to achieve noninvasive cancer treatment.
Collapse
Affiliation(s)
- Ming Yang
- Faculty of Materials Metallurgy and Chemistry, Jiangxi University of Science and Technology, Ganzhou 341000, P. R. China
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
| | - Wenzhi Ren
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| | - Haijing Cui
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
| | - Qiongyu Qin
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
| | - Qiuye Wang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
| | - Weihao Zhu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
| | - Xiaoxia Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
| | - Chunshu Pan
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
- Department of Radiology, Ningbo No. 2 Hospital, Ningbo 315010, P. R. China
| | - Xiaopeng Qi
- Faculty of Materials Metallurgy and Chemistry, Jiangxi University of Science and Technology, Ganzhou 341000, P. R. China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices and Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, P. R. China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China
| |
Collapse
|
20
|
Hajareh Haghighi F, Mercurio M, Cerra S, Salamone TA, Bianymotlagh R, Palocci C, Romano Spica V, Fratoddi I. Surface modification of TiO 2 nanoparticles with organic molecules and their biological applications. J Mater Chem B 2023; 11:2334-2366. [PMID: 36847384 DOI: 10.1039/d2tb02576k] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
In recent years, titanium(IV) dioxide nanoparticles (TiO2NPs) have shown promising potential in various biological applications such as antimicrobials, drug delivery, photodynamic therapy, biosensors, and tissue engineering. For employing TiO2NPs in these fields, their nanosurface must be coated or conjugated with organic and/or inorganic agents. This modification can improve their stability, photochemical properties, biocompatibility, and even surface area for further conjugation with other molecules such as drugs, targeting molecules, polymers, etc. This review describes the organic-based modification of TiO2NPs and their potential applications in the mentioned biological fields. In the first part of this review, around 75 recent publications (2017-2022) are mentioned on the common TiO2NP modifiers including organosilanes, polymers, small molecules, and hydrogels, which improve the photochemical features of TiO2NPs. In the second part of this review, we presented 149 recent papers (2020-2022) about the use of modified TiO2NPs in biological applications, in which specific bioactive modifiers are introduced in this part with their advantages. In this review, the following information is presented: (1) the common organic modifiers for TiO2NPs, (2) biologically important modifiers and their benefits, and (3) recent publications on biological studies on the modified TiO2NPs with their achievements. This review shows the paramount significance of the organic-based modification of TiO2NPs to enhance their biological effectiveness, paving the way toward the development of advanced TiO2-based nanomaterials in nanomedicine.
Collapse
Affiliation(s)
- Farid Hajareh Haghighi
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Martina Mercurio
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Sara Cerra
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | | | - Roya Bianymotlagh
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Cleofe Palocci
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy. .,Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Vincenzo Romano Spica
- Department of Movement, Health and Human Sciences, University of Rome Foro Italico, Piazza Lauro De Bosis, 15, 00135 Rome, Italy
| | - Ilaria Fratoddi
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
21
|
Yang XY, Lu YF, Xu JX, Du YZ, Yu RS. Recent Advances in Well-Designed Therapeutic Nanosystems for the Pancreatic Ductal Adenocarcinoma Treatment Dilemma. Molecules 2023; 28:molecules28031506. [PMID: 36771172 PMCID: PMC9920782 DOI: 10.3390/molecules28031506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with an extremely poor prognosis and low survival rate. Due to its inconspicuous symptoms, PDAC is difficult to diagnose early. Most patients are diagnosed in the middle and late stages, losing the opportunity for surgery. Chemotherapy is the main treatment in clinical practice and improves the survival of patients to some extent. However, the improved prognosis is associated with higher side effects, and the overall prognosis is far from satisfactory. In addition to resistance to chemotherapy, PDAC is significantly resistant to targeted therapy and immunotherapy. The failure of multiple treatment modalities indicates great dilemmas in treating PDAC, including high molecular heterogeneity, high drug resistance, an immunosuppressive microenvironment, and a dense matrix. Nanomedicine shows great potential to overcome the therapeutic barriers of PDAC. Through the careful design and rational modification of nanomaterials, multifunctional intelligent nanosystems can be obtained. These nanosystems can adapt to the environment's needs and compensate for conventional treatments' shortcomings. This review is focused on recent advances in the use of well-designed nanosystems in different therapeutic modalities to overcome the PDAC treatment dilemma, including a variety of novel therapeutic modalities. Finally, these nanosystems' bottlenecks in treating PDAC and the prospect of future clinical translation are briefly discussed.
Collapse
Affiliation(s)
- Xiao-Yan Yang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yuan-Fei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Jian-Xia Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, 318 Chaowang Road, Hangzhou 310005, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
- Correspondence: (Y.-Z.D.); (R.-S.Y.); Tel.: +86-571-88208435 (Y.-Z.D.); +86-571-87783925 (R.-S.Y.)
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
- Correspondence: (Y.-Z.D.); (R.-S.Y.); Tel.: +86-571-88208435 (Y.-Z.D.); +86-571-87783925 (R.-S.Y.)
| |
Collapse
|
22
|
Wang L, Song W, Choi S, Yu K, Zhang F, Guo W, Ma Y, Wang K, Qu F, Lin H. Hollow CoP@N-Carbon Nanospheres: Heterostructure and Glucose-Enhanced Charge Separation for Sonodynamic/Starvation Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2552-2563. [PMID: 36600575 DOI: 10.1021/acsami.2c15327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Sonodynamic therapy (SDT) can be described as ultrasonic (US) catalysis. Adequate charge separation is considered as effective means to promote reactive oxygen species (ROS). Here, hollow CoP@N-carbon@PEG (CPCs@PEG) nanospheres (∼60 nm) are prepared as sonosensitizers, showing greater ROS generation than pure CoP@PEG under US irradiation. Both 1O2 and ·O2- are activation species that are determined by O2 and electrons. The great SDT performance of CPCs@PEG is ascribed to the heterostructure which promotes the separation and transfer for US-generated electrons and holes. In addition, holes can be further captured by endogenous glucose that is in favor of electron aggregation and ROS generation. Moreover, the consumption of glucose would decrease intracellular ATP for starvation therapy. Given the higher oxidation ability of Co3+, CPCs@PEG nanospheres possess catalase (CAT) activity to convert H2O2 into O2 for assisting ROS generation. Moreover, they also can oxidize glutathione (GSH) as a mimic GSH oxidase to break intratumor redox balance, facilitating oxidative stress. More importantly, the nanocomposites reveal good degradation ability dominated by the oxidation from insoluble phosphide into soluble phosphate, accelerating elimination via urine and feces within 14 days. CPCs@PEG nanospheres integrate the above effects not only to reveal great tumor inhibition ability but also to excite immune activation for anticancer.
Collapse
Affiliation(s)
- Limin Wang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Wenhui Song
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Stephen Choi
- SXULTRASONIC (Shenzhen) LTD., Shenzhen518000, P.R. China
| | - Kai Yu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Feng Zhang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Wei Guo
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Yajie Ma
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Kai Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin150028, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin150028, China
| | - Fengyu Qu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
| | - Huiming Lin
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin150025, China
- Laboratory for Photon and Electronic Bandgap Materials, Ministry of Education, Harbin Normal University, Harbin150025, China
| |
Collapse
|