1
|
Zhu J, Li Y, Li X, Wang Y, Liu Q, Yang Y, Guan H. Feasibility analysis and development trend of nanomaterials for the treatment of pancreatic cancer. DISCOVER NANO 2025; 20:78. [PMID: 40354007 PMCID: PMC12069193 DOI: 10.1186/s11671-025-04259-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer is a highly aggressive disease that poses a significant threat to human health. Although conventional chemotherapy remains an effective treatment, it is often associated with severe side effects, underscoring the need for more effective cancer therapies. In this study, we analyzed the keywords of past studies, the countries with the highest number of publications, the leading journals, prominent authors, and collaborations between countries, authors, and journals, as well as the impact factors of relevant literature. The aim was to explore the trends in the use of nanomaterials for the treatment of pancreatic cancer, enabling researchers to review past achievements and gain a better understanding of future research directions. Relevant research articles were sourced from core Web of Science databases, and VOSviewer and CiteSpace visualization tools were employed to reveal the intrinsic links between the information. Research on the use of nanomaterials for the therapy of pancreatic cancer has been growing since the twenty-first century, particularly from 2018 to the present. The United States has become a leader in this field, with the highest number of publications and the most published authors. Additionally, a 2018 study published in Nature demonstrated that patients with insufficient CD8 + T-cell infiltration in the pancreatic cancer tumor microenvironment (TME) had significantly lower survival rates (HR = 2.5, p < 0.001). And CSF1R inhibitors combined with a PD-1 antibody resulted in 60% tumor shrinkage in a mouse model. These findings suggest that research on the tumor microenvironment and immunotherapy is poised to be a key focus of future studies, offering new hope for pancreatic cancer patients.
Collapse
Affiliation(s)
- Jingyao Zhu
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yue Li
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Xiaoqing Li
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yong Wang
- Stroke Center, Department of Neurology, Yanbian University Hospital (Yanbian Hospital), Ju Zi Road No.1327, Yanbian, 133000, Jilin, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Qixiang Liu
- Vascular Surgery Department of Jilin Provincial Vanguard Hospital, No. 1445 Qianjin Street, Chaoyang District, Changchun, China
| | - Yang Yang
- Stroke Center, Department of Neurology, Yanbian University Hospital (Yanbian Hospital), Ju Zi Road No.1327, Yanbian, 133000, Jilin, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - HongJian Guan
- Stroke Center, Department of Neurology, Yanbian University Hospital (Yanbian Hospital), Ju Zi Road No.1327, Yanbian, 133000, Jilin, China.
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China.
| |
Collapse
|
2
|
Huang X, Ji M, Shang X, Zhang H, Zhang X, Zhou J, Yin T. Smart on-demand drug release strategies for cancer combination therapy. J Control Release 2025; 383:113782. [PMID: 40294796 DOI: 10.1016/j.jconrel.2025.113782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
In cancer therapy, enhancing therapeutic indices and patient compliance has been a central focus in recent drug delivery technology development. However, achieving a delicate balance between improving anti-tumor efficacy and minimizing toxicity to normal tissues remains a significant challenge. With the advent of smart on-demand drug release strategies, new opportunities have emerged. These strategies represent a promising approach to drug delivery, enabling precise control over the release of therapeutic agents in a programmed and spatiotemporal manner. Recent studies have focused on designing delivery systems capable of releasing multiple therapeutic agents sequentially, while achieving spatial resolution in vivo. Smart on-demand drug release strategies have demonstrated considerable potential in tumor combination therapy for achieving precision drug delivery and controlled release by responding to specific physiological signals or external physical stimuli in the tumor microenvironment. These strategies not only improve tumor targeting and reduce toxicity to healthy tissues but also enable sequential release in combination therapy, allowing multiple drugs to be released in a specific spatiotemporal order to enhance synergistic treatment effects. In this paper, we systematically reviewed the current research progress of smart on-demand drug release drug delivery strategies in anti-tumor combination therapy. We highlighted representative integrated drug delivery systems and discussed the challenges associated with their clinical application. Additionally, potential future research directions are proposed to further advance this promising field.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Mengfei Ji
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xinyu Shang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hengchuan Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
3
|
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
|
4
|
Nair ST, Abhi C, Kamalasanan K, Pavithran K, Unni AR, Sithara MS, Sarma M, Mangalanandan TS. Pathophysiology-Driven Approaches for Overcoming Nanomedicine Resistance in Pancreatic Cancer. Mol Pharm 2024; 21:5960-5988. [PMID: 39561094 DOI: 10.1021/acs.molpharmaceut.4c00801] [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: 11/21/2024]
Abstract
Tumor heterogeneity poses a significant challenge in cancer therapy. To address this, we analyze pharmacotherapeutic challenges by categorizing them into static and dynamic barriers, reframing these challenges to improve drug delivery, efficacy, and the development of controlled-release nanomedicines (CRNMs). This pathophysiology-driven approach facilitates the design of novel therapeutics tailored to overcome obstacles in pancreatic ductal adenocarcinoma (PDAC) using nanotechnology. Advanced biomaterials in nanodrug delivery systems offer innovative solutions by combining controlled release, stimuli sensitivity, and smart design strategies. CRNMs are engineered to modulate spatiotemporal signaling and control drug release in PDAC, where resistance to conventional therapies is particularly high. This review explores pharmacokinetic considerations for nanomedicine design, RNA interference (RNAi) for stromal modulation, and the development of targeted nanomedicine strategies. Additionally, we highlight the limitations of current animal models in capturing the complexities of PDAC and discuss notable clinical failures, such as PEGylated hyaluronidase (Phase III HALO 109-301 trial) and evofosfamide (TH-302) with gemcitabine (MAESTRO trial), underscoring the need for improved models and treatment strategies. By targeting pathways like Notch and Hedgehog and incorporating stimuli-sensitive and pathway-modulating agents, CRNMs offer a promising avenue to enhance drug penetration and efficacy, reshaping the paradigm of pancreatic cancer treatment.
Collapse
Affiliation(s)
- Sreejith Thrivikraman Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - C Abhi
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Kaladhar Kamalasanan
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - K Pavithran
- Department of Medical Oncology and Hematology, School of Medicine, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Ashok R Unni
- Department of Veterinary Medicine, Central Animal Facility, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - M S Sithara
- Department of Veterinary Medicine, Central Animal Facility, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Manjit Sarma
- Department of Nuclear Medicine, Amrita School of Medicine, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - T S Mangalanandan
- Department of Endocrinology, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| |
Collapse
|
5
|
Luo W, Zhang T. The new era of pancreatic cancer treatment: Application of nanotechnology breaking through bottlenecks. Cancer Lett 2024; 594:216979. [PMID: 38795762 DOI: 10.1016/j.canlet.2024.216979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/02/2024] [Accepted: 05/16/2024] [Indexed: 05/28/2024]
Abstract
Since the advent of nanomedicine, physicians have harnessed these approaches for the prophylaxis, detection, and therapy of life-threatening diseases, particularly cancer. Nanoparticles have demonstrated notable efficacy in cancer therapy, showcasing the primary application of nanotechnology in targeted drug delivery. Pancreatic cancer stands out as the most lethal solid tumour in humans. The low survival rate is attributed to its highly aggressive nature, intrinsic resistance to chemotherapeutics, and the lack of successful therapies, compounded by delayed diagnosis due to nonspecific symptoms and the absence of rapid diagnostic strategies. Despite these challenges, nanotechnology-based carrier methods have been successfully employed in imaging and therapy approaches. Overcoming drug resistance in pancreatic cancer necessitates a comprehensive understanding of the microenvironment associated with the disease, paving the way for innovative nanocarriers. Hindered chemotherapy infiltration, attributed to inadequate vascularization and a dense tumour stroma, is a major hurdle that nanotechnology addresses. Intelligent delivery techniques, based on the Enhanced Permeability and Retention effect, form the basis of recently developed anticancer nanocarriers. These advancements aim to enhance drug accumulation in tumour locations, offering a potential solution to the treatment-resistant nature of cancer. Addressing the challenges in pancreatic cancer treatment demands innovative therapies, and the emergence of active nanocarriers presents a promising avenue for enhancing outcomes. This review specifically delves into the latest advancements in nanotechnology for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
6
|
Agarwal H, Bynum RC, Saleh N, Harris D, MacCuaig WM, Kim V, Sanderson EJ, Dennahy IS, Singh R, Behkam B, Gomez-Gutierrez JG, Jain A, Edil BH, McNally LR. Theranostic nanoparticles for detection and treatment of pancreatic cancer. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1983. [PMID: 39140128 PMCID: PMC11328968 DOI: 10.1002/wnan.1983] [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: 12/20/2023] [Revised: 06/21/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most recalcitrant cancers due to its late diagnosis, poor therapeutic response, and highly heterogeneous microenvironment. Nanotechnology has the potential to overcome some of the challenges to improve diagnostics and tumor-specific drug delivery but they have not been plausibly viable in clinical settings. The review focuses on active targeting strategies to enhance pancreatic tumor-specific uptake for nanoparticles. Additionally, this review highlights using actively targeted liposomes, micelles, gold nanoparticles, silica nanoparticles, and iron oxide nanoparticles to improve pancreatic tumor targeting. Active targeting of nanoparticles toward either differentially expressed receptors or PDAC tumor microenvironment (TME) using peptides, antibodies, small molecules, polysaccharides, and hormones has been presented. We focus on microenvironment-based hallmarks of PDAC and the potential for actively targeted nanoparticles to overcome the challenges presented in PDAC. It describes the use of nanoparticles as contrast agents for improved diagnosis and the delivery of chemotherapeutic agents that target various aspects within the TME of PDAC. Additionally, we review emerging nano-contrast agents detected using imaging-based technologies and the role of nanoparticles in energy-based treatments of PDAC. This article is categorized under: Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Happy Agarwal
- Stephenson Cancer Center, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Ryan C Bynum
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Nada Saleh
- Stephenson Cancer Center, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Danielle Harris
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - William M MacCuaig
- Stephenson Cancer Center, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Vung Kim
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Emma J Sanderson
- Stephenson Cancer Center, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Isabel S Dennahy
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Rohit Singh
- Stephenson Cancer Center, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech University, Blacksburg, Virginia, USA
| | | | - Ajay Jain
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Barish H Edil
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| | - Lacey R McNally
- Department of Surgery, University of Oklahoma Health Science, Oklahoma City, Oklahoma, USA
| |
Collapse
|
7
|
Zygmunt A, Gubernator J. Metabolism and structure of PDA as the target for new therapies: possibilities and limitations for nanotechnology. Expert Opin Drug Deliv 2024; 21:845-865. [PMID: 38899424 DOI: 10.1080/17425247.2024.2370492] [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: 01/23/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Certainly, pancreatic ductal adenocarcinoma poses one of the greatest challenges in current oncology. The dense extracellular matrix and low vessel density in PDA tumor impede the effective delivery of drugs, primarily due to the short pharmacokinetics of most drugs and potential electrostatic interactions with stroma components. AREA COVERED Owing to the distinctive metabolism of PDA and challenges in accessing nutrients, there is a growing interest in cell metabolism inhibitors as a potential means to inhibit cancer development. However, even if suitable combinations of inhibitors are identified, the question about their administration remains, as the same hindrances that impede effective treatment with conventional drugs will also hinder the delivery of inhibitors. Methods including nanotechnology to increase drugs in PDA penetrations are reviewed and discussed. EXPERT OPINION Pancreatic cancer is one of the most difficult tumors to treat due to the small number of blood vessels, high content of extracellular matrix, and specialized resistance mechanisms of tumor cells. One possible method of treating this tumor is the use of metabolic inhibitors in combinations that show synergy. Despite promising results in in vitro tests, their effect is uncertain due to the tumor's structure. In the case of pancreatic cancer, priming of the tumor tissue is required through the sequential administration of drugs that generate blood vessels, increase blood flow, and enhance vascular permeability and extracellular matrix. The use of drug carriers with a size of 10-30 nm may be crucial in the therapy of this cancer.
Collapse
Affiliation(s)
- Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
8
|
Wang L, Quine S, Frickenstein AN, Lee M, Yang W, Sheth VM, Bourlon MD, He Y, Lyu S, Garcia-Contreras L, Zhao YD, Wilhelm S. Exploring and Analyzing the Systemic Delivery Barriers for Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308446. [PMID: 38828467 PMCID: PMC11142462 DOI: 10.1002/adfm.202308446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 06/05/2024]
Abstract
Most nanomedicines require efficient in vivo delivery to elicit diagnostic and therapeutic effects. However, en route to their intended tissues, systemically administered nanoparticles often encounter delivery barriers. To describe these barriers, we propose the term "nanoparticle blood removal pathways" (NBRP), which summarizes the interactions between nanoparticles and the body's various cell-dependent and cell-independent blood clearance mechanisms. We reviewed nanoparticle design and biological modulation strategies to mitigate nanoparticle-NBRP interactions. As these interactions affect nanoparticle delivery, we studied the preclinical literature from 2011-2021 and analyzed nanoparticle blood circulation and organ biodistribution data. Our findings revealed that nanoparticle surface chemistry affected the in vivo behavior more than other nanoparticle design parameters. Combinatory biological-PEG surface modification improved the blood area under the curve by ~418%, with a decrease in liver accumulation of up to 47%. A greater understanding of nanoparticle-NBRP interactions and associated delivery trends will provide new nanoparticle design and biological modulation strategies for safer, more effective, and more efficient nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Skyler Quine
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Michael Lee
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit M. Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Margaret D. Bourlon
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lucila Garcia-Contreras
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73012, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma, 73019, USA
| |
Collapse
|
9
|
Zhong D, Cheng H, Liu H, Feng S, Liu Y, Xiang H, Chen J. Bibliometric analysis of Traditional Chinese Medicine nanoparticles research from 2005 to 2023. Electrophoresis 2024; 45:288-299. [PMID: 37909469 DOI: 10.1002/elps.202300207] [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: 09/19/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
To gain a deeper understanding of the current status of research on Traditional Chinese Medicine (TCM) and nanoparticles, we conducted a bibliometric study. We conducted a literature search in the Web of Science (WOS) for publications related to TCM and nanoparticles from 1992 to 2023. The data, including countries of publication, research institutions, journals, citations, and keywords, were analyzed using the Bibliometrix R-4.0 software package. We performed an analysis to identify the co-occurrence of keywords in the documents including their titles and abstracts. From 2005 to 2023, a total of 309 publications were included, with an average annual growth rate of 4.25%. The majority of these publications were published in Q1 journals (72, 47.06%) and Q2 journals (45, 29.41%). Among the 309 publications, 22 articles (7.12%) had an impact factor greater than 10, while 78 articles (25.24%) had an impact factor greater than 5. The analysis of international collaboration networks revealed limited international cooperation, with most collaborations occurring between institutions in China, the United States, and Australia. These 309 publications involved a total of 438 research institutions, with Chinese research institutions being the most prolific contributors. In this study, a total of 309 publications were included, comprising 1142 author keywords and 1175 keywords plus. Factor analysis of the 1175 keywords plus revealed that they could be grouped into five categories: one category included terms such as "oxide" and "zinc," another category included terms like "lipid" and "acid," a third category included terms such as "improve" and "enhance," a fourth category included terms like "silica" and "mesoporous," and the fifth category included terms like "PLGA" and "immune." Research on nanoparticles in TCM has been gradually gaining popularity. Currently, most of the research in this field is conducted in China, with limited international collaboration. The majority of TCM nanoparticle research focuses on individual herbal compounds, while research on nanoparticle formulations of traditional herbal prescriptions is relatively scarce.
Collapse
Affiliation(s)
- Dayuan Zhong
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
| | - Hui Cheng
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
- Institute of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Huixian Liu
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
- Post-graduate Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Shihui Feng
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
- Institute of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Yumei Liu
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
- Institute of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Huier Xiang
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
- Institute of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Jiaqi Chen
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, P. R. China
| |
Collapse
|
10
|
Li D, Chen X, Dai W, Jin Q, Wang D, Ji J, Tang BZ. Photo-Triggered Cascade Therapy: A NIR-II AIE Luminogen Collaborating with Nitric Oxide Facilitates Efficient Collagen Depletion for Boosting Pancreatic Cancer Phototheranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2306476. [PMID: 38157423 DOI: 10.1002/adma.202306476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/05/2023] [Indexed: 01/03/2024]
Abstract
The dense extracellular matrix (ECM) in the pancreatic cancer severely hampers the penetration of nanodrugs, which causes inferior therapeutic efficacy. To address this issue, a multifunctional liposome, namely, Lip-DTI/NO, integrating a type-I photosensitizer DTITBT with glutathione (GSH) or heat-responsive nitric oxide (NO) donor S-nitroso-N-acetyl-D-penicillamine (SNAP) is constructed to deplete the tumor ECM, leading to enhanced drug delivery and consequently improved phototherapy. The loaded DTITBT possesses multiple functions including NIR-II fluorescence imaging, efficient superoxide radical (O2 •- ) generation and excellent photothermal conversion efficiency, making it feasible for precisely pinpointing the tumor in the phototherapy process. Responding to the intracellular overexpressed glutathione or heat produced by photothermal effect of DTITBT, NO can be released from SNAP. Upon 808 nm laser irradiation, Lip-DTI/NO could selectively induce in situ generation of peroxynitrite anion (ONOO- ) in tumor after cascade processes including O2 •- production, GSH or heat-triggered NO release, and rapid reaction between O2 •- and NO. The generated ONOO- could activate the expression of endogenous matrix metalloproteinases which could efficiently digest collagen of tumor ECM, thus facilitating enhanced penetration and accumulation of Lip-DTI/NO in tumor. In vivo evaluation demonstrates the notable therapeutic efficacy via ONOO- -potentiated synergistic photodynamic-photothermal therapies on both subcutaneous and orthotopic pancreatic cancer model.
Collapse
Affiliation(s)
- Dan Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials College of Material Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xiaohui Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Wenbin Dai
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials College of Material Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, (CUHK-Shenzhen), Guangdong, 518172, China
| |
Collapse
|
11
|
Ding R, Li Y, Yu Y, Sun Z, Duan J. Prospects and hazards of silica nanoparticles: Biological impacts and implicated mechanisms. Biotechnol Adv 2023; 69:108277. [PMID: 37923235 DOI: 10.1016/j.biotechadv.2023.108277] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
With the thrive of nanotechnology, silica nanoparticles (SiNPs) have been extensively adopted in the agriculture, food, cosmetic, and even biomedical industries. Due to the mass production and use, SiNPs inevitably entered the environment, resulting in ecological toxicity and even posing a threat to human health. Although considerable investigations have been conducted to assess the toxicity of SiNPs, the correlation between SiNPs exposure and consequent health risks remains ambiguous. Since the biological impacts of SiNPs can differ from their design and application, the toxicity assessment for SiNPs may be extremely difficult. This review discussed the application of SiNPs in different fields, especially their biomedical use, and documented their potential release pathways into the environment. Meanwhile, the current process of assessing SiNPs-related toxicity on various model organisms and cell lines was also detailed, thus estimating the health threats posed by SiNPs exposure. Finally, the potential toxic mechanisms of SiNPs were also elaborated based on results obtained from both in vivo and in vitro trials. This review generally summarizes the biological effects of SiNPs, which will build up a comprehensive perspective of the application and toxicity of SiNPs.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
12
|
Silli EK, Li M, Shao Y, Zhang Y, Hou G, Du J, Liang J, Wang Y. Liposomal nanostructures for Gemcitabine and Paclitaxel delivery in pancreatic cancer. Eur J Pharm Biopharm 2023; 192:13-24. [PMID: 37758121 DOI: 10.1016/j.ejpb.2023.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/01/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Pancreatic cancer (PC) is an incurable disease with a high death rate in the world nowadays. Gemcitabine (GEM) and Paclitaxel (PTX) are considered as references of chemotherapeutic treatments and are commonly used in clinical applications. Factors related to the tumor microenvironment such as insufficient tumor penetration, toxicity, and drug resistance can limit the effectiveness of these therapeutic anticancer drugs. The use of different liposomal nanostructures is a way that can optimize the drug's effectiveness and reduce toxicity. Given the development of PC therapy, this review focuses on advances in Nano-formulation, characterization, and delivery systems of loaded GEM and PTX liposomes using chemotherapy, nucleic acid delivery, and stroma remodeling therapy. As a result, the review covers the literature dealing with the applications of liposomes in PC therapy.
Collapse
Affiliation(s)
- Epiphane K Silli
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Mengfei Li
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Yuting Shao
- College of Engineering, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Yiran Zhang
- College of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Guilin Hou
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jiaqian Du
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jingdan Liang
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ying Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
13
|
Bellapu KK, Joga R, Kannan BR, Yerram S, Varpe P, Mergu T, Vasu PY, Srivastava S, Kumar S. Lipid-based mesoporous silica nanoparticles: a paradigm shift in management of pancreatic cancer. Pharm Pat Anal 2023; 12:261-273. [PMID: 38214201 DOI: 10.4155/ppa-2023-0024] [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: 01/13/2024]
Abstract
Pancreatic adenocarcinoma, a devastating disease, has the worst cancer prognosis in humans. It often develops resistance to common chemotherapy medications, such as gemcitabine, taxol and 5-fluorouracil. The dense stroma limits therapeutic efficacy in treating this disease. Low or limited drug loading capacity is another problem with current chemotherapeutic agents. There is a need to develop novel approaches to overcome these issues. Hence, an innovative approach has been proposed to co-deliver both hydrophilic (Gemcitabine) and hydrophobic (Paclitaxel) drugs in a single carrier using lipid bilayer-mesoporous silica nanoparticles (LB-MSNP). MSNPs offer effective drug delivery due to their superior bioavailability and physicochemical properties. Further, in order to achieve clinical translation and regulatory approval, toxicity and biodegradability of MSNPs must be resolved.
Collapse
Affiliation(s)
- Kiran Kumar Bellapu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Bharthi R Kannan
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Sravani Yerram
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Priya Varpe
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Tejaswini Mergu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Pavan Y Vasu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Saurabh Srivastava
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| |
Collapse
|
14
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
15
|
Raza F, Evans L, Motallebi M, Zafar H, Pereira-Silva M, Saleem K, Peixoto D, Rahdar A, Sharifi E, Veiga F, Hoskins C, Paiva-Santos AC. Liposome-based diagnostic and therapeutic applications for pancreatic cancer. Acta Biomater 2023; 157:1-23. [PMID: 36521673 DOI: 10.1016/j.actbio.2022.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the harshest and most challenging cancers to treat, often labeled as incurable. Chemotherapy continues to be the most popular treatment yet yields a very poor prognosis. The main barriers such as inefficient drug penetration and drug resistance, have led to the development of drug carrier systems. The benefits, ease of fabrication and modification of liposomes render them as ideal future drug delivery systems. This review delves into the versatility of liposomes to achieve various mechanisms of treatment for pancreatic cancer. Not only are there benefits of loading chemotherapy drugs and targeting agents onto liposomes, as well as mRNA combined therapy, but liposomes have also been exploited for immunotherapy and can be programmed to respond to photothermal therapy. Multifunctional liposomal formulations have demonstrated significant pre-clinical success. Functionalising drug-encapsulated liposomes has resulted in triggered drug release, specific targeting, and remodeling of the tumor environment. Suppressing tumor progression has been achieved, due to their ability to more efficiently and precisely deliver chemotherapy. Currently, no multifunctional surface-modified liposomes are clinically approved for pancreatic cancer thus we aim to shed light on the trials and tribulations and progress so far, with the hope for liposomal therapy in the future and improved patient outcomes. STATEMENT OF SIGNIFICANCE: Considering that conventional treatments for pancreatic cancer are highly associated with sub-optimal performance and systemic toxicity, the development of novel therapeutic strategies holds outmost relevance for pancreatic cancer management. Liposomes are being increasingly considered as promising nanocarriers for providing not only an early diagnosis but also effective, highly specific, and safer treatment, improving overall patient outcome. This manuscript is the first in the last 10 years that revises the advances in the application of liposome-based formulations in bioimaging, chemotherapy, phototherapy, immunotherapy, combination therapies, and emergent therapies for pancreatic cancer management. Prospective insights are provided regarding several advantages resulting from the use of liposome technology in precision strategies, fostering new ideas for next-generation diagnosis and targeted therapies of pancreatic cancer.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Lauren Evans
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Mahzad Motallebi
- Immunology Board for Transplantation And Cell-based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 7616911319, Iran; Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Kalsoom Saleem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 45320, Pakistan
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Clare Hoskins
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
16
|
Ahmad A, Rashid S, Chaudhary AA, Alawam AS, Alghonaim MI, Raza SS, Khan R. Nanomedicine as potential cancer therapy via targeting dysregulated transcription factors. Semin Cancer Biol 2023; 89:38-60. [PMID: 36669712 DOI: 10.1016/j.semcancer.2023.01.002] [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: 09/30/2022] [Revised: 01/02/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Cancer as a disease possess quite complicated pathophysiological implications and is among the prominent causes of morbidity and mortality on global scales. Anti-cancer chemotherapy, surgery, and radiation therapy are some of the present-day conventional treatment options. However, these therapeutic paradigms own several retreats, including lack of specificity, non-targeted toxicological implications, inefficient drug delivery to targeted cells, and emergence of cancer resistance, ultimately causing ineffective cancer management. Owing to the advanced and better biophysical characteristic features and potentiality for the tailoring and customizations and in several fashions, nanotechnology can entirely transubstantiate the cancer identification and its managements. Additionally, nanotechnology also renders several answers to present-day mainstream limitations springing-up in anti-cancer therapeutics. Nanocarriers, owing to their outstanding physicochemical features including but not limited to their particle size, surface morphological features viz. shape etc., have been employed in nanomedicinal platforms for targeting various transcription factors leading to worthy pharmacological outcomes. This transcription targeting activates the wide array of cellular and molecular events like antioxidant enzyme-induction, apoptotic cell death, cell-cycle arrest etc. These outcomes are obtained after the activation or inactivation of several transcription factors and cellular pathways. Further, nanoformulations have been precisely calibrated and functionalized with peculiar targeting groups for improving their efficiency to deliver the drug-payload to specified and targeted cancerous cells and tissues. This review undertakes an extensive, across-the-board and all-inclusive approach consisting of various studies encompassing different types of tailored and customized nanoformulations and nanomaterials designed for targeting the transcription factors implicated in the process of carcinogenesis, tumor-maturation, growth and metastasis. Various transcription factors viz. nuclear factor kappa (NF-κB), signal transducer and activators of transcription (STAT), Cmyc and Twist-related protein 1 (TWIST1) along with several types of nanoparticles targeting these transcription factors have been summarized here. A section has also been dedicated to the different types of nanoparticles targeting the hypoxia inducing factors. Efforts have been made to summarize several other transcription factors implicated in various stages of cancer development, growth, progression and invasion, and their targeting with different kinds of nanomedicinal agents.
Collapse
Affiliation(s)
- Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Ibrahim Alghonaim
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Syed Shadab Raza
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow 226003, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| |
Collapse
|
17
|
Cheng Y, Zheng X, Zhang L, Zhao J, Hu L, Wang S. Enhanced photothermal and chemotherapy of pancreatic tumors by degrading the extracellular matrix. Colloids Surf B Biointerfaces 2023; 221:113010. [PMID: 36375292 DOI: 10.1016/j.colsurfb.2022.113010] [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: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
The degradation of extracellular matrix (ECM) to increase drug permeability is an attractive approach to enhancing pancreatic cancer therapy efficiency. Herein, polypyrrole nanoparticles (PPy NPs) were prepared by a template-guided chemical oxidation method. These PPy NPs with abundant surface pores were used to load the anticancer drug doxorubicin (DOX). In order to intelligently control the DOX release, PPy/DOX NPs were further entrapped with a thermoresponsive ligand, lauric acid (LA), to form PPy-LA/DOX NPs. Bromelain (BL) was then grafted onto the surface of PPy-LA NPs or PPy-LA/DOX NPs through an amidation reaction with the carboxyl group of LA. It was found that the DOX release of PPy-LA/DOX NPs was pH and temperature responsive, reaching a maximum amount of 85.9% within 48 h at pH = 5.4 and 50 °C. Moreover, it was demonstrated that the resultant PLB (PPy-LA-BL) NPs could efficiently hydrolyze the collagen in ECM and enhance the permeability of DOX to the pancreatic tumor. Remarkably, PLB NPs not only featured admirable photothermal conversion but also exhibited obvious photoacoustic imaging capability, which enabled imaging-guided enhanced tumor ablation. This study is anticipated to provide a feasible strategy to improve the permeability of nanoparticles to tumors.
Collapse
Affiliation(s)
- Yuancun Cheng
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Xiaoyi Zheng
- Department of Gastroenterology, Changhai Hospital, Noval Military Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Liying Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Noval Military Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Lianghao Hu
- Department of Gastroenterology, Changhai Hospital, Noval Military Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China.
| |
Collapse
|
18
|
Borges R, Pelosine AM, de Souza ACS, Machado J, Justo GZ, Gamarra LF, Marchi J. Bioactive Glasses as Carriers of Cancer-Targeted Drugs: Challenges and Opportunities in Bone Cancer Treatment. MATERIALS (BASEL, SWITZERLAND) 2022; 15:9082. [PMID: 36556893 PMCID: PMC9781635 DOI: 10.3390/ma15249082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 05/20/2023]
Abstract
The treatment of bone cancer involves tumor resection followed by bone reconstruction of the defect caused by the tumor using biomaterials. Additionally, post-surgery protocols cover chemotherapy, radiotherapy, or drug administration, which are employed as adjuvant treatments to prevent tumor recurrence. In this work, we reviewed new strategies for bone cancer treatment based on bioactive glasses as carriers of cancer-targeted and other drugs that are intended for bone regeneration in conjunction with adjuvant treatments. Drugs used in combination with bioactive glasses can be classified into cancer-target, osteoclast-target, and new therapies (such as gene delivery and bioinorganic). Microparticulated, nanoparticulated, or mesoporous bioactive glasses have been used as drug-delivery systems. Additionally, surface modification through functionalization or the production of composites based on polymers and hydrogels has been employed to improve drug-release kinetics. Overall, although different drugs and drug delivery systems have been developed, there is still room for new studies involving kinase inhibitors or antibody-conjugated drugs, as these drugs have been poorly explored in combination with bioactive glasses.
Collapse
Affiliation(s)
- Roger Borges
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André 09210-580, Brazil
| | - Agatha Maria Pelosine
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André 09210-580, Brazil
| | | | - Joel Machado
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo, Diadema 05508-070, Brazil
| | - Giselle Zenker Justo
- Departamento de Bioquímica, Universidade Federal de São Paulo, São Paulo 05508-070, Brazil
| | | | - Juliana Marchi
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André 09210-580, Brazil
| |
Collapse
|
19
|
Najafi M, Tavakol S, Zarrabi A, Ashrafizadeh M. Dual role of quercetin in enhancing the efficacy of cisplatin in chemotherapy and protection against its side effects: a review. Arch Physiol Biochem 2022; 128:1438-1452. [PMID: 32521182 DOI: 10.1080/13813455.2020.1773864] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemotherapy has opened a new window in cancer therapy. However, the resistance of cancer cells has dramatically reduced the efficacy of chemotherapy. Cisplatin is a chemotherapeutic agent and its potential in cancer therapy has been restricted by resistance of cancer cells. As a consequence, the scientists have attempted to find new strategies in elevating chemotherapy efficacy. Due to great anti-tumour activity, naturally occurring compounds are of interest in polychemotherapy. Quercetin is a flavonoid with high anti-tumour activity against different cancers that can be used with cisplatin to enhance its efficacy and also are seen to sensitise cancer cells into chemotherapy. Furthermore, cisplatin has side effects such as nephrotoxicity and ototoxicity. Administration of quercetin is advantageous in reducing the adverse effects of cisplatin without compromising its anti-tumour activity. In this review, we investigate the dual role of quercetin in enhancing anti-tumour activity of cisplatin and simultaneous reduction in its adverse effects.
Collapse
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Turkey
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
20
|
Sun R, Xiang J, Zhou Q, Piao Y, Tang J, Shao S, Zhou Z, Bae YH, Shen Y. The tumor EPR effect for cancer drug delivery: Current status, limitations, and alternatives. Adv Drug Deliv Rev 2022; 191:114614. [PMID: 36347432 DOI: 10.1016/j.addr.2022.114614] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2022]
Abstract
Over the past three decades, the enhanced permeability and retention (EPR) effect has been considered the basis of tumor-targeted drug delivery. Various cancer nanomedicines, including macromolecular drugs, have been designed to utilize this mechanism for preferential extravasation and accumulation in solid tumors. However, such nanomedicines have not yet achieved convincing therapeutic benefits in clinics. Increasing evidence suggests that the EPR effect is over-represented in human tumors, especially in metastatic tumors. This review covers the evolution of the concept, the heterogeneity and limitation of the EPR effect in clinical realities, and prospects for alternative strategies independent of the EPR effect.
Collapse
Affiliation(s)
- Rui Sun
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - You Han Bae
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
21
|
Jiang Z, Zhang W, Zhang J, Liu T, Xing J, Zhang H, Tang D. Nanomaterial-Based Drug Delivery Systems: A New Weapon for Cancer Immunotherapy. Int J Nanomedicine 2022; 17:4677-4696. [PMID: 36211025 PMCID: PMC9541303 DOI: 10.2147/ijn.s376216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer immunotherapy, a major breakthrough in cancer treatment, has been successfully applied to treat a number of tumors. However, given the presence of factors in the tumor microenvironment (TME) that impede immunotherapy, only a small proportion of patients achieve a good clinical response. With the ability to increase permeability and cross biological barriers, nanomaterials have been successfully applied to deliver immunotherapeutic agents, thus realizing the anti-cancer therapeutic potential of therapeutic agents. This has driven a wave of research into systems for the delivery of immunotherapeutic agents, which has resulted in widespread interest in nanomaterial-based drug delivery systems. Nanomaterial-based drug delivery systems are able to overcome the challenges from TME and thus achieve good results in cancer immunotherapy. If it can make a breakthrough in improving biocompatibility and reducing cytotoxicity, it will be more widely used in clinical practice. Different types of nanomaterials may also have some subtle differences in enhancing cancer immunotherapy. Moreover, delivery systems made of nanomaterials loaded with drugs, such as cytotoxic drugs, cytokines, and adjuvants, could be used for cancer immunotherapy because they avoid the toxicity and side effects associated with these drugs, thereby enabling their reuse. Therefore, further insights into nanomaterial-based drug delivery systems will provide more effective treatment options for cancer patients.
Collapse
Affiliation(s)
- Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Tian Liu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| |
Collapse
|
22
|
Noubissi Nzeteu GA, Gibbs BF, Kotnik N, Troja A, Bockhorn M, Meyer NH. Nanoparticle-based immunotherapy of pancreatic cancer. Front Mol Biosci 2022; 9:948898. [PMID: 36106025 PMCID: PMC9465485 DOI: 10.3389/fmolb.2022.948898] [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: 05/20/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) has a complex and unique tumor microenvironment (TME). Due to the physical barrier formed by the desmoplastic stroma, the delivery of drugs to the tumor tissue is limited. The TME also contributes to resistance to various immunotherapies such as cancer vaccines, chimeric antigen receptor T cell therapy and immune checkpoint inhibitors. Overcoming and/or modulating the TME is therefore one of the greatest challenges in developing new therapeutic strategies for PC. Nanoparticles have been successfully used as drug carriers and delivery systems in cancer therapy. Recent experimental and engineering developments in nanotechnology have resulted in increased drug delivery and improved immunotherapy for PC. In this review we discuss and analyze the current nanoparticle-based immunotherapy approaches that are at the verge of clinical application. Particularly, we focus on nanoparticle-based delivery systems that improve the effectiveness of PC immunotherapy. We also highlight current clinical research that will help to develop new therapeutic strategies for PC and especially targeted immunotherapies based on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Gaetan Aime Noubissi Nzeteu
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
- *Correspondence: N. Helge Meyer, ; Gaetan Aime Noubissi Nzeteu,
| | - Bernhard F. Gibbs
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Nika Kotnik
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Achim Troja
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - Maximilian Bockhorn
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - N. Helge Meyer
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
- *Correspondence: N. Helge Meyer, ; Gaetan Aime Noubissi Nzeteu,
| |
Collapse
|
23
|
Smart pH-responsive polyhydralazine/bortezomib nanoparticles for remodeling tumor microenvironment and enhancing chemotherapy. Biomaterials 2022; 288:121737. [PMID: 36031455 DOI: 10.1016/j.biomaterials.2022.121737] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/01/2022] [Accepted: 08/06/2022] [Indexed: 01/01/2023]
Abstract
The clinical translation of nanomedicines has been impeded by the unfavorable tumor microenvironment (TME), particularly the tortuous vasculature networks, which significantly influence the transport and distribution of nanomedicines into tumors. In this work, a smart pH-responsive bortezomib (BTZ)-loaded polyhydralazine nanoparticle (PHDZ/BTZ) is presented, which has a great capacity to augment the accumulation of BTZ in tumors by dilating tumor blood vessels via specific release of vasodilator hydralazine (HDZ). The Lewis acid-base coordination effect between the boronic bond of BTZ and amino of HDZ empowered PHDZ/BTZ nanoparticles with great stability and high drug loading contents. Once triggered by the acidic tumor environment, HDZ could be released quickly to remodel TME through tumor vessel dilation, hypoxia attenuation, and lead to an increased intratumoral BTZ accumulation. Additionally, our investigation revealed that this pH-responsive nanoparticle dramatically suppressed tumor growth, inhibited the occurrence of lung metastasis with fewer side effects and induced immunogenic cell death (ICD), thereby eliciting immune activation including massive cytotoxic T lymphocytes (CTLs) infiltration in tumors and efficient serum proinflammatory cytokine secretion compared with free BTZ treatment. Thus, with efficient drug loading capacity and potent immune activation, PHDZ nanoparticles exhibit great potential in the delivery of boronic acid-containing drugs aimed at a wide range of diseases.
Collapse
|
24
|
Ni N, Wang W, Sun Y, Sun X, Leong DT. Inducible endothelial leakiness in nanotherapeutic applications. Biomaterials 2022; 287:121640. [PMID: 35772348 DOI: 10.1016/j.biomaterials.2022.121640] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/02/2022]
Abstract
All intravenous delivered nanomedicine needs to escape from the blood vessel to exert their therapeutic efficacy at their designated site of action. Failure to do so increases the possibility of detrimental side effects and negates their therapeutic intent. Many powerful anticancer nanomedicine strategies rely solely on the tumor derived enhanced permeability and retention (EPR) effect for the only mode of escaping from the tumor vasculature. However, not all tumors have the EPR effect nor can the EPR effect be induced or controlled for its location and timeliness. In recent years, there have been exciting developments along the lines of inducing endothelial leakiness at the tumor to decrease the dependence of EPR. Physical disruption of the endothelial-endothelial cell junctions with coordinated biological intrinsic pathways have been proposed that includes various modalities like ultrasound, radiotherapy, heat and even nanoparticles, appear to show good progress towards the goal of inducing endothelial leakiness. This review explains the intricate and complex biological background behind the endothelial cells with linkages on how updated reported nanomedicine strategies managed to induce endothelial leakiness. This review will also end off with fresh insights on where the future of inducible endothelial leakiness holds.
Collapse
Affiliation(s)
- Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Weiyi Wang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yu Sun
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore; Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, PR China
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.
| |
Collapse
|
25
|
Gillson J, Abd El-Aziz YS, Leck LYW, Jansson PJ, Pavlakis N, Samra JS, Mittal A, Sahni S. Autophagy: A Key Player in Pancreatic Cancer Progression and a Potential Drug Target. Cancers (Basel) 2022; 14:3528. [PMID: 35884592 PMCID: PMC9315706 DOI: 10.3390/cancers14143528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer is known to have the lowest survival outcomes among all major cancers, and unfortunately, this has only been marginally improved over last four decades. The innate characteristics of pancreatic cancer include an aggressive and fast-growing nature from powerful driver mutations, a highly defensive tumor microenvironment and the upregulation of advantageous survival pathways such as autophagy. Autophagy involves targeted degradation of proteins and organelles to provide a secondary source of cellular supplies to maintain cell growth. Elevated autophagic activity in pancreatic cancer is recognized as a major survival pathway as it provides a plethora of support for tumors by supplying vital resources, maintaining tumour survival under the stressful microenvironment and promoting other pathways involved in tumour progression and metastasis. The combination of these features is unique to pancreatic cancer and present significant resistance to chemotherapeutic strategies, thus, indicating a need for further investigation into therapies targeting this crucial pathway. This review will outline the autophagy pathway and its regulation, in addition to the genetic landscape and tumor microenvironment that contribute to pancreatic cancer severity. Moreover, this review will also discuss the mechanisms of novel therapeutic strategies that inhibit autophagy and how they could be used to suppress tumor progression.
Collapse
Affiliation(s)
- Josef Gillson
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
| | - Yomna S. Abd El-Aziz
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Lionel Y. W. Leck
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Patric J. Jansson
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Nick Pavlakis
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
| | - Jaswinder S. Samra
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
| | - Anubhav Mittal
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
- School of Medicine, University of Notre Dame, Darlinghurst, Sydney, NSW 2010, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
| |
Collapse
|
26
|
Huo T, Zhang X, Qian M, Nie H, Liang D, Lin C, Yang Y, Guo W, Lächelt U, Huang R. A Space-Time Conversion Vehicle for Programmed Multi-Drugs Delivery into Pancreatic Tumor to Overcome Matrix and Reflux Barriers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200608. [PMID: 35508899 PMCID: PMC9284157 DOI: 10.1002/advs.202200608] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/28/2022] [Indexed: 05/15/2023]
Abstract
The numerous biological barriers, which limit pharmacotherapy of pancreatic carcinoma, including inadequate drug accumulation in the tumor environment, a dense extracellular matrix (ECM) and efficient drug-efflux mechanisms, illustrate the requirement of multifunctional delivery systems to overcome the individual barriers at the right place at the right time. Herein, a space-time conversion vehicle based on covalent organic framework (COF)-coated mesoporous silica nanospheres (MSN) with a sandwiched polyethyleneimine (PEI) layer (MPCP), is designed. The space-specific drugs-loaded vehicle (MG PP CL P) is obtained by separately incorporating a chemotherapeutic agent (gemcitabine, G) into the MSN core, a P glycoprotein inhibitor (LY 335979, P) into the PEI layer, and an extracellular matrix disruptor (losartan, L) into the COF shell. Thereafter, a programmed drug delivery is achieved via the ordered degradation from COF shell to MSN core. Sequential release of the individual drugs, synergized with a change of nanoparticle surface charge, contribute to an obvious extracellular matrix distraction, distinct drug efflux inhibition, and consequently enhance chemotherapeutic outcomes in pancreatic carcinoma. This MPCP-based vehicle design suggests a robust space-time conversion strategy to achieve programmed multi-drugs delivery and represents a new avenue to the treatment of pancreatic carcinoma by overcoming extracellular matrix and drug reflux barriers.
Collapse
Affiliation(s)
- Taotao Huo
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Xiaoyi Zhang
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Min Qian
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Huifang Nie
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Dong Liang
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Chenteng Lin
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Yafeng Yang
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Wei Guo
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| | - Ulrich Lächelt
- Department of Pharmaceutical SciencesUniversity of ViennaVienna1090Austria
| | - Rongqin Huang
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryMinistry of EducationFudan UniversityShanghai201203P. R. China
| |
Collapse
|
27
|
Tarannum M, Holtzman K, Dréau D, Mukherjee P, Vivero-Escoto JL. Nanoparticle combination for precise stroma modulation and improved delivery for pancreatic cancer. J Control Release 2022; 347:425-434. [PMID: 35569588 DOI: 10.1016/j.jconrel.2022.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
Therapeutic success in the treatment of pancreatic ductal adenocarcinoma (PDAC) is hindered by the extensive stroma associated to this disease. Stroma is composed of cellular and non-cellular components supporting and evolving with the tumor. One of the most studied mediators of cancer cell-stroma crosstalk is sonic hedgehog (SHh) pathway leading to the intense desmoplasia observed in PDAC tumors. Herein, we demonstrate that the use of mesoporous silica nanoparticles (MSNs) containing an SHh inhibitor, cyclopamine (CyP), and the combination of chemotherapeutic drugs (Gemcitabine (Gem)/cisplatin (cisPt)) as the main delivery system for the sequential treatment led to the reduction in tumor stroma along with an improvement in the treatment of PDAC. We synthesized two versions of the MSN-based platform containing the SHh inhibitor (CyP-MSNs) and the drug combination (PEG-Gem-cisPt-MSNs). In vitro and in vivo protein analysis show that CyP-MSNs effectively inhibited the SHh pathway. In addition, the sequential combination of CyP-MSNs followed by PEG-Gem-cisPt-MSNs led to effective stromal modulation, increased access of secondary PEG-Gem-cisPt-MSNs at the tumor site, and improved therapeutic performance in HPAF II xenograft mice. Taken together, our findings support the potential of drug delivery using MSNs for stroma modulation and to prevent pancreatic cancer progression.
Collapse
Affiliation(s)
- Mubin Tarannum
- Department of Chemistry, University of North Carolina Charlotte, Charlotte, NC 28223, USA; Nanoscale Science Program, University of North Carolina Charlotte, Charlotte, NC 28223, USA
| | - Katherine Holtzman
- Department of Biological Sciences, University of North Carolina Charlotte, Charlotte, NC 28223, USA
| | - Didier Dréau
- Department of Biological Sciences, University of North Carolina Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina Charlotte, Charlotte, NC 28223, USA
| | - Pinku Mukherjee
- Department of Biological Sciences, University of North Carolina Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina Charlotte, Charlotte, NC 28223, USA
| | - Juan L Vivero-Escoto
- Department of Chemistry, University of North Carolina Charlotte, Charlotte, NC 28223, USA; Nanoscale Science Program, University of North Carolina Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
28
|
Munir MU. Nanomedicine Penetration to Tumor: Challenges, and Advanced Strategies to Tackle This Issue. Cancers (Basel) 2022; 14:cancers14122904. [PMID: 35740570 PMCID: PMC9221319 DOI: 10.3390/cancers14122904] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Nanomedicine has been under investigation for several years to improve the efficiency of chemotherapeutics, having minimal pharmacological effects clinically. Ineffective tumor penetration is mediated by tumor environments, including limited vascular system, rising cancer cells, higher interstitial pressure, and extra-cellular matrix, among other things. Thus far, numerous methods to increase nanomedicine access to tumors have been described, including the manipulation of tumor micro-environments and the improvement of nanomedicine characteristics; however, such outdated approaches still have shortcomings. Multi-functional convertible nanocarriers have recently been developed as an innovative nanomedicine generation with excellent tumor infiltration abilities, such as tumor-penetrating peptide-mediated transcellular transport. The developments and limitations of nanomedicines, as well as expectations for better outcomes of tumor penetration, are discussed in this review.
Collapse
Affiliation(s)
- Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| |
Collapse
|
29
|
Tarannum M, Vivero-Escoto JL. Nanoparticle-based therapeutic strategies targeting major clinical challenges in pancreatic cancer treatment. Adv Drug Deliv Rev 2022; 187:114357. [PMID: 35605679 DOI: 10.1016/j.addr.2022.114357] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers due to its aggressiveness and the challenges for early diagnosis and treatment. Recently, nanotechnology has demonstrated relevant strategies to overcome some of the major clinical issues in the treatment of PDAC. This review is focused on the pathological hallmarks of PDAC and the impact of nanotechnology to find solutions. It describes the use of nanoparticle-based systems designed for the delivery of chemotherapeutic agents and combinatorial alternatives that address the chemoresistance associated with PDAC, the development of combination therapies targeting the molecular heterogeneity in PDAC, the investigation of novel therapies dealing with the improvement of immunotherapy and handling the desmoplastic stroma in PDAC by remodeling the tumor microenvironment. A special section is dedicated to the design of nanoparticles for unique non-traditional modalities that could be promising in the future for the improvement in the dismal prognosis of PDAC.
Collapse
|
30
|
Nel AE, Mei KC, Liao YP, Lu X. Multifunctional Lipid Bilayer Nanocarriers for Cancer Immunotherapy in Heterogeneous Tumor Microenvironments, Combining Immunogenic Cell Death Stimuli with Immune Modulatory Drugs. ACS NANO 2022; 16:5184-5232. [PMID: 35348320 PMCID: PMC9519818 DOI: 10.1021/acsnano.2c01252] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In addition to the contribution of cancer cells, the solid tumor microenvironment (TME) has a critical role in determining tumor expansion, antitumor immunity, and the response to immunotherapy. Understanding the details of the complex interplay between cancer cells and components of the TME provides an unprecedented opportunity to explore combination therapy for intervening in the immune landscape to improve immunotherapy outcome. One approach is the introduction of multifunctional nanocarriers, capable of delivering drug combinations that provide immunogenic stimuli for improvement of tumor antigen presentation, contemporaneous with the delivery of coformulated drug or synthetic molecules that provide immune danger signals or interfere in immune-escape, immune-suppressive, and T-cell exclusion pathways. This forward-looking review will discuss the use of lipid-bilayer-encapsulated liposomes and mesoporous silica nanoparticles for combination immunotherapy of the heterogeneous immune landscapes in pancreatic ductal adenocarcinoma and triple-negative breast cancer. We describe how the combination of remote drug loading and lipid bilayer encapsulation is used for the synthesis of synergistic drug combinations that induce immunogenic cell death, interfere in the PD-1/PD-L1 axis, inhibit the indoleamine-pyrrole 2,3-dioxygenase (IDO-1) immune metabolic pathway, restore spatial access to activated T-cells to the cancer site, or reduce the impact of immunosuppressive stromal components. We show how an integration of current knowledge and future discovery can be used for a rational approach to nanoenabled cancer immunotherapy.
Collapse
Affiliation(s)
- André E. Nel
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, United States
| | - Kuo-Ching Mei
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Lu
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
31
|
Fu Y, Saraswat AL, Monpara J, Patel K. Stromal disruption facilitating invasion of a 'nano-arsenal' into the solid tumor. Drug Discov Today 2022; 27:1132-1141. [PMID: 34823002 PMCID: PMC12045031 DOI: 10.1016/j.drudis.2021.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/01/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Owing to the indispensable role of nanotechnology in cancer therapy, it is imperative to comprehend every aspect limiting its therapeutic potential. Several preclinical reports have demonstrated the enhanced permeability and retention (EPR)-mediated preferential tumor uptake of nanoparticles. However, the therapeutic outcome of nanotherapeutics is severely compromised by heterogeneous drug distribution and insufficient penetration of nanomedicine in a solid tumor owing to the dense tumor extracellular matrix (ECM). Herein, we elaborate on various preclinically investigated tumor stromal disrupting strategies, which we call 'cannons', to compromise the impenetrable 'fortress-like' solid tumor microenvironment. We have described and summarized major approaches to enhance the penetration of a 'nano-arsenal' in solid tumors. ECM remodeling strategies could be very beneficial in enhancing the therapeutic efficacy of monoclonal antibodies and translational nanomedicine.
Collapse
Affiliation(s)
- Yige Fu
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Genentech, Inc, South San Francisco, CA, USA
| | - Aishwarya L Saraswat
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | | | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
32
|
Peng H, Shen J, Long X, Zhou X, Zhang J, Xu X, Huang T, Xu H, Sun S, Li C, Lei P, Wu H, Zhao J. Local Release of TGF-β Inhibitor Modulates Tumor-Associated Neutrophils and Enhances Pancreatic Cancer Response to Combined Irreversible Electroporation and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105240. [PMID: 35128843 PMCID: PMC8981446 DOI: 10.1002/advs.202105240] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/11/2022] [Indexed: 05/09/2023]
Abstract
Pancreatic cancer is a deadly disease with little response to standard therapies. Irreversible electroporation (IRE) has emerged as a novel ablative technique for the clinical treatment of pancreatic cancer. Combinations of IRE and immunotherapies, including anti-programmed death 1 (αPD1) immune checkpoint blockade, have shown promising efficacy in both preclinical and clinical studies. However, tumor recurrence remains an obstacle that needs to be overcome. It herein is shown that IRE induces a substantial infiltration of neutrophils into pancreatic tumors. These neutrophils are then polarized into a protumor phenotype by immunosuppressive cues, in particular transforming growth factor β (TGF-β). Using glutathione-responsive degradable mesoporous silica nanoparticles loaded with SB525334, an inhibitor of TGF-β1 receptor, it is demonstrated that local inhibition of TGF-β within the tumor microenvironment promotes neutrophil polarization into an antitumor phenotype, enhances pancreatic cancer response to combined IRE and αPD1 therapy, and induces long-term antitumor memory. The therapeutic efficacy is also attributed to tumor infiltration by CD8+ cytotoxic T cells, depletion of regulatory T cells, and maturation of antigen-presenting dendritic cells. Thus, modulating neutrophil polarization with nanomedicine is a promising strategy for treating pancreatic cancer.
Collapse
Affiliation(s)
- Huiming Peng
- Department of AnatomySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Jian Shen
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430022China
| | - Xin Long
- Department of Histology and EmbryologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Xiaoqi Zhou
- Department of ImmunologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Jiaqi Zhang
- Department of AnatomySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Xina Xu
- Department of AnatomySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Teng Huang
- Department of AnatomySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Hui Xu
- Ultrastructural Pathology LaboratoryDepartment of PathologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Shuguo Sun
- Department of AnatomySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Chun Li
- Department of Cancer Systems ImagingUniversity of Texas MD Anderson CancerHoustonTX77030USA
| | - Ping Lei
- Department of ImmunologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Heshui Wu
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430022China
| | - Jun Zhao
- Department of AnatomySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubei Province430030China
- Department of Nuclear Medicine and PETTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
- Cell Architecture Research CenterHuazhong University of Science and TechnologyWuhanHubei Province430030China
| |
Collapse
|
33
|
Slapak EJ, el Mandili M, Bijlsma MF, Spek CA. Mesoporous Silica Nanoparticle-Based Drug Delivery Systems for the Treatment of Pancreatic Cancer: A Systematic Literature Overview. Pharmaceutics 2022; 14:390. [PMID: 35214121 PMCID: PMC8876630 DOI: 10.3390/pharmaceutics14020390] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is a devastating disease with the worst outcome of any human cancer. Despite significant improvements in cancer treatment in general, little progress has been made in pancreatic cancer (PDAC), resulting in an overall 5-year survival rate of less than 10%. This dismal prognosis can be attributed to the limited clinical efficacy of systemic chemotherapy due to its high toxicity and consequent dose reductions. Targeted delivery of chemotherapeutic drugs to PDAC cells without affecting healthy non-tumor cells will largely reduce collateral toxicity leading to reduced morbidity and an increased number of PDAC patients eligible for chemotherapy treatment. To achieve targeted delivery in PDAC, several strategies have been explored over the last years, and especially the use of mesoporous silica nanoparticles (MSNs) seem an attractive approach. MSNs show high biocompatibility, are relatively easy to surface modify, and the porous structure of MSNs enables high drug-loading capacity. In the current systematic review, we explore the suitability of MSN-based targeted therapies in the setting of PDAC. We provide an extensive overview of MSN-formulations employed in preclinical PDAC models and conclude that MSN-based tumor-targeting strategies may indeed hold therapeutic potential for PDAC, although true clinical translation has lagged behind.
Collapse
Affiliation(s)
- Etienne J. Slapak
- Center of Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.e.M.); (C.A.S.)
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
- Oncode Institute, 1105 AZ Amsterdam, The Netherlands
| | - Mouad el Mandili
- Center of Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.e.M.); (C.A.S.)
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
| | - Maarten F. Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands;
- Oncode Institute, 1105 AZ Amsterdam, The Netherlands
| | - C. Arnold Spek
- Center of Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.e.M.); (C.A.S.)
| |
Collapse
|
34
|
Abstract
Applications of AIEgens in biosensing, disease diagnosis, and drug release.
Collapse
Affiliation(s)
- Guangfu Feng
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, P.R. China
| | - Sijie Liao
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, P.R. China
| | - Yufeng Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, P.R. China
| | - Huaizu Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, P.R. China
| | - Xingyu Luo
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, P.R. China
| | - Xiangming Zhou
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, P.R. China
| | - Jun Fang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, P.R. China
| |
Collapse
|
35
|
Han H, Li S, Zhong Y, Huang Y, Wang K, Jin Q, Ji J, Yao K. Emerging pro-drug and nano-drug strategies for gemcitabine-based cancer therapy. Asian J Pharm Sci 2022; 17:35-52. [PMID: 35261643 PMCID: PMC8888143 DOI: 10.1016/j.ajps.2021.06.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/19/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Gemcitabine has been extensively applied in treating various solid tumors. Nonetheless, the clinical performance of gemcitabine is severely restricted by its unsatisfactory pharmacokinetic parameters and easy deactivation mainly because of its rapid deamination, deficiencies in deoxycytidine kinase (DCK), and alterations in nucleoside transporter. On this account, repeated injections with a high concentration of gemcitabine are adopted, leading to severe systemic toxicity to healthy cells. Accordingly, it is highly crucial to fabricate efficient gemcitabine delivery systems to obtain improved therapeutic efficacy of gemcitabine. A large number of gemcitabine pro-drugs were synthesized by chemical modification of gemcitabine to improve its biostability and bioavailability. Besides, gemcitabine-loaded nano-drugs were prepared to improve the delivery efficiency. In this review article, we introduced different strategies for improving the therapeutic performance of gemcitabine by the fabrication of pro-drugs and nano-drugs. We hope this review will provide new insight into the rational design of gemcitabine-based delivery strategies for enhanced cancer therapy.
Collapse
Affiliation(s)
- Haijie Han
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Su Li
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yueyang Zhong
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Kai Wang
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
36
|
Li J, Xie Q, Ma R, Li Y, Yuan J, Ren M, Li H, Wang J, Lu D, Xu Z, Wang J. Recent Progress on the Synergistic Antitumor Effect of a Borneol-Modified Nanocarrier Drug Delivery System. Front Med (Lausanne) 2021; 8:750170. [PMID: 34901063 PMCID: PMC8655685 DOI: 10.3389/fmed.2021.750170] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023] Open
Abstract
Borneol, a traditional Chinese medicine, can enhance therapeutic efficacy by guiding the active ingredients to the target site. Reportedly, borneol improves the penetration capacity of the nasal, cornea, transdermal, intestinal, and blood-brain barriers. Although nanotechnology dramatically changed the face of oncology by targeting tumor sites, the efficiency of nanoparticles delivered to tumor sites is very low, with only 0.7% of the total particles delivered. Thus, based on the penetration ability and the inhibition drug efflux of borneol, it was expected to increase the targeting and detention efficacy of drugs into tumor sites in nanocarriers with borneol modification. Borneol modified nanocarriers used to improve drug-targeting has become a research focus in recent years, but few studies in this area, especially in the antitumor application. Hence, this review summarizes the recent development of nanocarriers with borneol modification. We focus on the updated works of improving therapeutic efficacy, reducing toxicity, inhibiting tumor metastasis, reversing multidrug resistance, and enhancing brain targeting to expand their application and provide a reference for further exploration of targeting drug delivery systems for solid tumor treatment.
Collapse
Affiliation(s)
- Jinxiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianmei Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Danni Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
37
|
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
|
38
|
de Lázaro I, Mooney DJ. Obstacles and opportunities in a forward vision for cancer nanomedicine. NATURE MATERIALS 2021; 20:1469-1479. [PMID: 34226688 DOI: 10.1038/s41563-021-01047-7] [Citation(s) in RCA: 238] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/01/2021] [Indexed: 05/14/2023]
Abstract
Cancer nanomedicines were initially envisioned as magic bullets, travelling through the circulation to target tumours while sparing healthy tissues the toxicity of classic chemotherapy. While a limited number of nanomedicine therapies have resulted, the disappointing news is that major obstacles were overlooked in the nanoparticle's journey. However, some of these challenges may be turned into opportunities. Here, we discuss biological barriers to cancer nanomedicines and elaborate on two directions that the field is currently exploring to meet its initial expectations. The first strategy entails re-engineering cancer nanomedicines to prevent undesired interactions en route to the tumour. The second aims instead to leverage these obstacles into out-of-the-box diagnostic and therapeutic applications of nanomedicines, for cancer and beyond. Both paths require, among other developments, a deeper understanding of nano-bio interactions. We offer a forward look at how classic cancer nanomedicine may overcome its limitations while contributing to other areas of research.
Collapse
Affiliation(s)
- Irene de Lázaro
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
| |
Collapse
|
39
|
Efficient nano-enabled therapy for gastrointestinal cancer using silicasome delivery technology. Sci China Chem 2021. [DOI: 10.1007/s11426-021-1126-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
40
|
Chen TT, Yuan MM, Tao YM, Ren XY, Li S. Engineering of Self-assembly Polymers Encapsulated with Dual Anticancer Drugs for the Treatment of Endometrial Cancer. J CLUST SCI 2021. [DOI: 10.1007/s10876-021-02175-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Igarashi K, Cabral H, Hong T, Anraku Y, Mpekris F, Stylianopoulos T, Khan T, Matsumoto A, Kataoka K, Matsumoto Y, Yamasoba T. Vascular Bursts Act as a Versatile Tumor Vessel Permeation Route for Blood-Borne Particles and Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103751. [PMID: 34528759 DOI: 10.1002/smll.202103751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Indexed: 06/13/2023]
Abstract
Dynamic bursting in tumor vasculature has recently sparked interest as a novel particle transportation route for drug delivery. These bursts facilitate the transport of sub-100 nm nanoparticles into tumors, though their contribution on the access of other blood-borne particles remains unknown. To evaluate the versatility of this phenomenon, the in vivo kinetics of a variety of intravenously injected particles and their penetration in tumor xenografts and allografts are compared. Dextran, polymeric micelles, liposomes, and polymeric vesicles with diameters ranging from 32 to 302 nm are found to colocalize in virtually all vascular bursts. By mathematical modeling, the burst vent size is estimated to be 625 nm or larger, indicating the dynamic and stochastic formation of large permeation routes in tumor vasculature. Furthermore, some burst vents are found to be µm-sized, allowing the transport of 1 µm microspheres. Moreover, antibody drugs and platelets are capable of utilizing vascular burst transportation, demonstrating the application of this phenomenon to other types of therapeutics and cellular components. These findings indicate the vast potential of vascular bursts, extending the biological and therapeutic significance of this phenomenon to a wide range of blood-borne particles and cells.
Collapse
Affiliation(s)
- Kazunori Igarashi
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Taehun Hong
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, 1678, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, 1678, Cyprus
| | - Thahomina Khan
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Akira Matsumoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
- Kanagawa Institute of Industrial Science and Technology, 3-25-13 Tono-machi, Kawasaki-ku, Kawasaki City, Kanagawa, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tono-machi, Kawasaki-ku, Kawasaki City, Kanagawa, 210-0821, Japan
- Institute of Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 133-0033, Japan
| | - Yu Matsumoto
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
42
|
He B, Sui X, Yu B, Wang S, Shen Y, Cong H. Recent advances in drug delivery systems for enhancing drug penetration into tumors. Drug Deliv 2021; 27:1474-1490. [PMID: 33100061 PMCID: PMC7594734 DOI: 10.1080/10717544.2020.1831106] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The emergence of nanomaterials for drug delivery provides the opportunity to avoid the side effects of systemic drug administration and injury caused by the removal of tumors, delivering great promise for future cancer treatments. However, the efficacy of current nano drugs is not significantly better than that of the original drug treatments. The important reason is that nano drugs enter the tumor vasculature, remaining close to the blood vessels and unable to enter the tumor tissue or tumor cells to complete the drug delivery process. The low efficiency of drug penetration into tumors has become a bottleneck restricting the development of nano-drugs. Herein, we present a systematic overview of recent advances on the design of nano-drug carriers in drug delivery systems for enhancing drug penetration into tumors. The review is organized into four sections: The drug penetration process in tumor tissue includes paracellular and transcellular transport, which is summarized first. Strategies that promote tumor penetration are then introduced, including methods of remodeling the tumor microenvironment, charge inversion, dimensional change, and surface modification of ligands which promote tissue penetration. Conclusion and the prospects for the future development of drug penetration are finally briefly illustrated. The review is intended to provide thoughts for effective treatment of cancer by summarizing strategies for promoting the endocytosis of nano drugs into tumor cells.
Collapse
Affiliation(s)
- Bin He
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin Sui
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Key Laboratory of Bio-Fibers and Eco-Textiles, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Key Laboratory of Bio-Fibers and Eco-Textiles, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Immunological effects of nano-enabled hyperthermia for solid tumors: opportunity and challenge. Front Chem Sci Eng 2021. [DOI: 10.1007/s11705-021-2059-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
44
|
Chen J, Li S, Liu X, Liu S, Xiao C, Zhang Z, Li S, Li Z, Yang X. Transforming growth factor-β blockade modulates tumor mechanical microenvironments for enhanced antitumor efficacy of photodynamic therapy. NANOSCALE 2021; 13:9989-10001. [PMID: 34076013 DOI: 10.1039/d1nr01552d] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Photodynamic therapy (PDT) is frequently used in cancer treatment in clinical settings. However, its applications in stroma-rich solid tumors, e.g., triple negative breast cancer, are limited by abnormal mechanical microenvironments. Solid stress accumulated in stroma-rich solid tumors compresses tumor blood vessels, hampers the delivery of photosensitizers (PSs) in tumor tissues, and poses a major challenge for potent PDT. Here, we report a novel combination strategy to augment PDT based cancer therapy by combining hydroxyethyl starch-chlorin e6 conjugate self-assembled nanoparticles (HES-Ce6 NPs) with the transforming growth factor-β (TGFβ) inhibitor LY2157299 (LY). HES-Ce6 conjugates, as synthesized by one step esterification reaction, could self-assemble into uniform HES-Ce6 NPs, which exhibited enhanced photostability and generated more reactive oxygen species (ROS) under 660 nm laser irradiation than free Ce6. Prior to PDT, intragastric administration of LY decreased collagen deposition, alleviated solid stress, and decompressed tumor blood vessels. As a result, the reconstructed tumor mechanical microenvironment promoted accumulation and penetration of HES-Ce6 NPs into tumor tissues, contributing to augmented antitumor efficacy of HES-Ce6 NP mediated PDT. Modulating tumor mechanical microenvironments using TGFβ blockade to enhance the delivery of PSs in tumors with excessive extracellular matrix represents an efficient strategy for treating stroma-rich solid tumors.
Collapse
Affiliation(s)
- Jitang Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang Y, Gao Z, Du X, Chen S, Zhang W, Wang J, Li H, He X, Cao J, Wang J. Co-inhibition of the TGF-β pathway and the PD-L1 checkpoint by pH-responsive clustered nanoparticles for pancreatic cancer microenvironment regulation and anti-tumor immunotherapy. Biomater Sci 2021; 8:5121-5132. [PMID: 32820750 DOI: 10.1039/d0bm00916d] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dense extracellular matrix (ECM) surrounding tumor cells to sequester CD8+ T cell infiltration and prevent drug penetration. Concomitant inhibition of both the TGF-β pathway and the PD-1/PD-L1 checkpoint is a viable strategy to increase T cell infiltration and cytotoxicity. Here, we used an acidic tumor extracellular pH (pHe) responsive clustered nanoparticle (LYiClustersiPD-L1) to deliver TGF-β receptor inhibitors (LY2157299) and siRNA targeting PD-L1 (siPD-L1) for PDAC stroma microenvironment regulation and antitumor immunotherapy. LY2157299 encapsulated in the hydrophobic core of the nanoparticle can effectively inhibit the activation of pancreatic stellate cells (PSCs) and result in a reduction in type I collagen. siPD-L1 adsorbed on the surface of the nanoparticle was released with small size poly(amidoamine) (PAMAM) at the surface of LYiClustersiPD-L1 under pHe and penetrated into the tumors to silence PD-L1 gene expression in tumor cells. Compared to monotherapy, LYiClustersiPD-L1 significantly increased tumor infiltrating CD8+ T cells and provoked antitumor immunity to synergistically suppress tumor growth in both a subcutaneous Panc02 xenograft model and an orthotopic tumor model.
Collapse
Affiliation(s)
- Yang Wang
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China. and Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zhuxin Gao
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China. and Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xiaojiao Du
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China. and Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Senbiao Chen
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Wangcheng Zhang
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jilong Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Hongjun Li
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xinyu He
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jie Cao
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China. and Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China. and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China and Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China and Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, P.R. China
| |
Collapse
|
46
|
Liang T, Zhang B, Xing Z, Dong Y, Xu H, Chen X, Jiang L, Zhu J, Min Q. Adapting and Remolding: Orchestrating Tumor Microenvironment Normalization with Photodynamic Therapy by Size Transformable Nanoframeworks. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102180] [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]
Affiliation(s)
- Tingxizi Liang
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Benhua Zhang
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Zejing Xing
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Yuxiang Dong
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Hongmei Xu
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Xueqin Chen
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Liping Jiang
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Jun‐Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science Chemistry and Biomedicine Innovation Center School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| |
Collapse
|
47
|
Liang T, Zhang B, Xing Z, Dong Y, Xu H, Chen X, Jiang L, Zhu JJ, Min Q. Adapting and Remolding: Orchestrating Tumor Microenvironment Normalization with Photodynamic Therapy by Size Transformable Nanoframeworks. Angew Chem Int Ed Engl 2021; 60:11464-11473. [PMID: 33751758 DOI: 10.1002/anie.202102180] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 12/11/2022]
Abstract
Abnormal tumor microenvironment (TME) facilitates tumor proliferation and metastasis and establishes physiological barriers for effective transport of therapeutics inside the tumor, posing great challenges for cancer treatment. We designed a core-satellite size transformable nanoframework (denoted as T-PFRT) that can synchronously adapt to and remold TME for augmenting photodynamic therapy to inhibit tumor growth and prevent tumor metastasis. Upon matrix metalloproteinase 2 (MMP2)-responsive dissociation of the nanoframework in TME, the core structure loaded with TGFβ signaling pathway inhibitor and oxygen-carrying hemoglobin aims to stroma remodeling and hypoxia relief, allowing photosensitizer-encapsulated satellite particles to penetrate to deep-seated tumor for oxygen-fueled photodynamic therapy. T-PFRT could overcome the stroma and hypoxia barriers for delivering therapeutics and gain excellent therapeutic outcomes in the treatment of primary and metastatic tumors.
Collapse
Affiliation(s)
- Tingxizi Liang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Benhua Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zejing Xing
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yuxiang Dong
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Hongmei Xu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xueqin Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Liping Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
48
|
Liu Y, Zhou J, Li Q, Li L, Jia Y, Geng F, Zhou J, Yin T. Tumor microenvironment remodeling-based penetration strategies to amplify nanodrug accessibility to tumor parenchyma. Adv Drug Deliv Rev 2021; 172:80-103. [PMID: 33705874 DOI: 10.1016/j.addr.2021.02.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Remarkable advances in nano delivery systems have provided new hope for tumor prevention, diagnosis and treatment. However, only limited clinical therapeutic effects against solid tumors were achieved. One of the main reasons is the presence of abundant physiological and pathological barriers in vivo that impair tumoral penetration and distribution of the nanodrugs. These barriers are related to the components of tumor microenvironment (TME) including abnormal tumor vasculature, rich composition of the extracellular matrix (ECM), and abundant stroma cells. Herein, we review the advanced strategies of TME remodeling to overcome these biological obstacles against nanodrug delivery. This review aims to offer a perspective guideline for the implementation of promising approaches to facilitate intratumoral permeation of nanodrugs through alleviation of biological barriers. At the same time, we analyze the advantages and disadvantages of the corresponding methods and put forward possible directions for the future researches.
Collapse
Affiliation(s)
- Yanhong Liu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jiyuan Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Qiang Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Lingchao Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yue Jia
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Feiyang Geng
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
49
|
Hu X, Xia F, Lee J, Li F, Lu X, Zhuo X, Nie G, Ling D. Tailor-Made Nanomaterials for Diagnosis and Therapy of Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002545. [PMID: 33854877 PMCID: PMC8025024 DOI: 10.1002/advs.202002545] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/25/2020] [Indexed: 05/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide due to its aggressiveness and the challenge to early diagnosis and treatment. In recent decades, nanomaterials have received increasing attention for diagnosis and therapy of PDAC. However, these designs are mainly focused on the macroscopic tumor therapeutic effect, while the crucial nano-bio interactions in the heterogeneous microenvironment of PDAC remain poorly understood. As a result, the majority of potent nanomedicines show limited performance in ameliorating PDAC in clinical translation. Therefore, exploiting the unique nature of the PDAC by detecting potential biomarkers together with a deep understanding of nano-bio interactions that occur in the tumor microenvironment is pivotal to the design of PDAC-tailored effective nanomedicine. This review will introduce tailor-made nanomaterials-enabled laboratory tests and advanced noninvasive imaging technologies for early and accurate diagnosis of PDAC. Moreover, the fabrication of a myriad of tailor-made nanomaterials for various PDAC therapeutic modalities will be reviewed. Furthermore, much preferred theranostic multifunctional nanomaterials for imaging-guided therapies of PDAC will be elaborated. Lastly, the prospects of these nanomaterials in terms of clinical translation and potential breakthroughs will be briefly discussed.
Collapse
Affiliation(s)
- Xi Hu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Fan Xia
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Jiyoung Lee
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Fangyuan Li
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| | - Xiaoyang Lu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Xiaozhen Zhuo
- Department of Cardiologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyNo.11 Zhongguancun BeiyitiaoBeijing100190China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510700China
| | - Daishun Ling
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| |
Collapse
|
50
|
Peter AE, Sandeep BV, Rao BG, Kalpana VL. Nanotechnology to the Rescue: Treatment Perspective for the Immune Dysregulation Observed in COVID-19. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.644023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The study of the use of nanotechnology for drug delivery has been extensive. Nanomedical approaches for therapeutics; drug delivery in particular is superior to conventional methods in that it allows for controlled targeted delivery and release, higher stability, extended circulation time, minimal side-effects, and improved pharmacokinetic clearance (of the drug) form the body, to name a few. The magnitude of COVID-19, the current ongoing pandemic has been severe; it has caused widespread the loss of human life. In individuals with severe COVID-19, immune dysregulation and a rampant state of hyperinflammation is observed. This kind of an immunopathological response is detrimental and results in rapid disease progression, development of secondary infections, sepsis and can be fatal. Several studies have pin-pointed the reason for this immune dysregulation; deviations in the signaling pathways involved in the mediation and control of immune responses. In severe COVID-19 patients, many signaling cascades including JAK/STAT, NF-κB, MAPK/ERK, TGF beta, VEGF, and Notch signaling were found to be either upregulated or inactivated. Targeting these aberrant signaling pathways in conjunction with antiviral therapy will effectuate mitigation of the hyperinflammation, hypercytokinemia, and promote faster recovery. The science of the use of nanocarriers as delivery agents to modulate these signaling pathways is not new; it has already been explored for other inflammatory diseases and in particular, cancer therapy. Numerous studies have evaluated the efficacy and potential of nanomedical approaches to modulate these signaling pathways and have been met with positive results. A treatment regime, that includes nanotherapeutics and antiviral therapies will prove effective and holds great promise for the successful treatment of COVID-19. In this article, we review different nanomedical approaches already studied for targeting aberrant signaling pathways, the host immune response to SARS-CoV-2, immunopathology and the dysregulated signaling pathways observed in severe COVID-19 and the current treatment methods in use for targeting signaling cascades in COVID-19. We then conclude by suggesting that the use of nanomedical drug delivery systems for targeting signaling pathways can be extended to effectively target the aberrant signaling pathways in COVID-19 for best treatment results.
Collapse
|