1
|
Zhang Z, Wang R, Chen L. Drug Delivery System Targeting Cancer-Associated Fibroblast for Improving Immunotherapy. Int J Nanomedicine 2025; 20:483-503. [PMID: 39816375 PMCID: PMC11734509 DOI: 10.2147/ijn.s500591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of non-malignant cells that play a crucial role in the tumor microenvironment, increasingly recognized as key contributors to cancer progression, metastasis, and treatment resistance. So, targeting CAFs has always been considered an important part of cancer immunotherapy. However, targeting CAFs to improve the efficacy of tumor therapy is currently a major challenge. Nanomaterials show their unique advantages in the whole process. At present, nanomaterials have achieved significant accomplishments in medical applications, particularly in the field of cancer-targeted therapy, showing enormous potential. It has been confirmed that nanomaterials can not only directly target CAFs, but also interact with the tumor microenvironment (TME) and immune cells to affect tumorigenesis. As for the cancer treatment, nanomaterials could enhance the therapeutic effect in many ways. Therefore, in this review, we first summarized the current understanding of the complex interactions between CAFs and TME, immune cells, and tumor cells. Next, we discussed common nanomaterials in modern medicine and their respective impacts on the TME, CAFs, and interactions with tumors. Finally, we focus on the application of nano drug delivery system targeting CAFs in cancer therapy.
Collapse
Affiliation(s)
- Zhongsong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Rong Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| |
Collapse
|
2
|
Wu C, Zhai Y, Ji J, Yang X, Ye L, Lu G, Shi X, Zhai G. Advances in tumor stroma-based targeted delivery. Int J Pharm 2024; 664:124580. [PMID: 39142464 DOI: 10.1016/j.ijpharm.2024.124580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The tumor stroma plays a crucial role in tumor progression, and the interactions between the extracellular matrix, tumor cells, and stromal cells collectively influence tumor progression and the efficacy of therapeutic agents. Currently, utilizing components of the tumor stroma for drug delivery is a noteworthy strategy. A number of targeted drug delivery systems designed based on tumor stromal components are entering clinical trials. Therefore, this paper provides a thorough examination of the function of tumor stroma in the advancement of targeted drug delivery systems. One approach is to use tumor stromal components for targeted drug delivery, which includes certain stromal components possessing inherent targeting capabilities like HA, laminin, along with targeting stromal cells homologously. Another method entails directly focusing on tumor stromal components to reshape the tumor stroma and facilitate drug delivery. These drug delivery systems exhibit great potential in more effective cancer therapy strategies, such as precise targeting, enhanced penetration, improved safety profile, and biocompatibility. Ultimately, the deployment of these drug delivery systems can deepen our comprehension of tumor stroma and the advanced development of corresponding drug delivery systems.
Collapse
Affiliation(s)
- Chunyan Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
3
|
Wells K, Liu T, Zhu L, Yang L. Immunomodulatory nanoparticles activate cytotoxic T cells for enhancement of the effect of cancer immunotherapy. NANOSCALE 2024; 16:17699-17722. [PMID: 39257225 DOI: 10.1039/d4nr01780c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Cancer immunotherapy represents a promising targeted treatment by leveraging the patient's immune system or adoptive transfer of active immune cells to selectively eliminate cancer cells. Despite notable clinical successes, conventional immunotherapies face significant challenges stemming from the poor infiltration of endogenous or adoptively transferred cytotoxic T cells in tumors, immunosuppressive tumor microenvironment and the immune evasion capability of cancer cells, leading to limited efficacy in many types of solid tumors. Overcoming these hurdles is essential to broaden the applicability of immunotherapies. Recent advances in nanotherapeutics have emerged as an innovative tool to overcome these challenges and enhance the therapeutic potential of tumor immunotherapy. The unique biochemical and biophysical properties of nanomaterials offer advantages in activation of immune cells in vitro for cell therapy, targeted delivery, and controlled release of immunomodulatory agents in vivo. Nanoparticles are excellent carriers for tumor associated antigens or neoantigen peptides for tumor vaccine, empowering activation of tumor specific T cell responses. By precisely delivering immunomodulatory agents to the tumor site, immunoactivating nanoparticles can promote tumor infiltration of endogenous T cells or adoptively transferred T cells into tumors, to overcoming delivery and biological barriers in the tumor microenvironment, augmenting the immune system's ability to recognize and eliminate cancer cells. This review provides an overview of the current advances in immunotherapeutic approaches utilizing nanotechnology. With a focus on discussions concerning strategies to enhance activity and efficacy of cytotoxic T cells and explore the intersection of engineering nanoparticles and immunomodulation aimed at bolstering T cell-mediated immune responses, we introduce various nanoparticle formulations designed to deliver therapeutic payloads, tumor antigens and immunomodulatory agents for T cell activation. Diverse mechanisms through which nanoparticle-based approaches influence T cell responses by improving antigen presentation, promoting immune cell trafficking, and reprogramming immunosuppressive tumor microenvironments to potentiate anti-tumor immunity are examined. Additionally, the synergistic potential of combining nanotherapeutics with existing immunotherapies, such as immune checkpoint inhibitors and adoptive T cell therapies is explored. In conclusion, this review highlights emerging research advances on activation of cytotoxic T cells using nanoparticle agents to support the promises and potential applications of nanoparticle-based immunomodulatory agents for cancer immunotherapy.
Collapse
Affiliation(s)
- Kory Wells
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tongrui Liu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lei Zhu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
4
|
Abbasifard M, Bagherzadeh K, Khorramdelazad H. The story of clobenpropit and CXCR4: can be an effective drug in cancer and autoimmune diseases? Front Pharmacol 2024; 15:1410104. [PMID: 39070795 PMCID: PMC11272485 DOI: 10.3389/fphar.2024.1410104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Clobenpropit is a histamine H3 receptor antagonist and has developed as a potential therapeutic drug due to its ability to inhibit CXCR4, a chemokine receptor involved in autoimmune diseases and cancer pathogenesis. The CXCL12/CXCR4 axis involves several biological phenomena, including cell proliferation, migration, angiogenesis, inflammation, and metastasis. Accordingly, inhibiting CXCR4 can have promising clinical outcomes in patients with malignancy or autoimmune disorders. Based on available knowledge, Clobenpropit can effectively regulate the release of monocyte-derived inflammatory cytokine in autoimmune diseases such as juvenile idiopathic arthritis (JIA), presenting a potential targeted target with possible advantages over current therapeutic approaches. This review summarizes the intricate interplay between Clobenpropit and CXCR4 and the molecular mechanisms underlying their interactions, comprehensively analyzing their impact on immune regulation. Furthermore, we discuss preclinical and clinical investigations highlighting the probable efficacy of Clobenpropit for managing autoimmune diseases and cancer. Through this study, we aim to clarify the immunomodulatory role of Clobenpropit and its advantages and disadvantages as a novel therapeutic opportunity.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Kowsar Bagherzadeh
- Eye Research Center, The Five Senses Health Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
5
|
Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, Liu X, Ba X, Xia D, Peng E, Chen Z, Tang K. The Application of Nanoparticles Targeting Cancer-Associated Fibroblasts. Int J Nanomedicine 2024; 19:3333-3365. [PMID: 38617796 PMCID: PMC11012801 DOI: 10.2147/ijn.s447350] [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: 11/12/2023] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAF) are the most abundant stromal cells in the tumor microenvironment (TME), especially in solid tumors. It has been confirmed that it can not only interact with tumor cells to promote cancer progression and metastasis, but also affect the infiltration and function of immune cells to induce chemotherapy and immunotherapy resistance. So, targeting CAF has been considered an important method in cancer treatment. The rapid development of nanotechnology provides a good perspective to improve the efficiency of targeting CAF. At present, more and more researches have focused on the application of nanoparticles (NPs) in targeting CAF. These studies explored the effects of different types of NPs on CAF and the multifunctional nanomedicines that can eliminate CAF are able to enhance the EPR effect which facilitate the anti-tumor effect of themselves. There also exist amounts of studies focusing on using NPs to inhibit the activation and function of CAF to improve the therapeutic efficacy. The application of NPs targeting CAF needs to be based on an understanding of CAF biology. Therefore, in this review, we first summarized the latest progress of CAF biology, then discussed the types of CAF-targeting NPs and the main strategies in the current. The aim is to elucidate the application of NPs in targeting CAF and provide new insights for engineering nanomedicine to enhance immune response in cancer treatment.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
6
|
Yu Z, Huang L, Guo J. Anti-stromal nanotherapeutics for hepatocellular carcinoma. J Control Release 2024; 367:500-514. [PMID: 38278367 DOI: 10.1016/j.jconrel.2024.01.050] [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: 10/27/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Hepatocellular carcinoma (HCC), the most commonly diagnosed primary liver cancer, has become a leading cause of cancer-related death worldwide. Accumulating evidence confirms that the stromal constituents within the tumor microenvironment (TME) exacerbate HCC malignancy and set the barriers to current anti-HCC treatments. Recent developments of nano drug delivery system (NDDS) have facilitated the application of stroma-targeting therapeutics, disrupting the stromal TME in HCC. This review discusses the stromal activities in HCC development and therapy resistance. In addition, it addresses the delivery challenges of NDDS for stroma-targeting therapeutics (termed anti-stromal nanotherapeutics in this review), and provides recent advances in anti-stromal nanotherapeutics for safe, effective, and specific HCC therapy.
Collapse
Affiliation(s)
- Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
7
|
Schwartz-Duval A, Mackeyev Y, Mahmud I, Lorenzi PL, Gagea M, Krishnan S, Sokolov KV. Intratumoral Biosynthesis of Gold Nanoclusters by Pancreatic Cancer to Overcome Delivery Barriers to Radiosensitization. ACS NANO 2024; 18:1865-1881. [PMID: 38206058 PMCID: PMC10811688 DOI: 10.1021/acsnano.3c04260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024]
Abstract
Nanoparticle delivery to solid tumors is a prime challenge in nanomedicine. Here, we approach this challenge through the lens of biogeochemistry, the field that studies the flow of chemical elements within ecosystems as manipulated by living cellular organisms and their environments. We leverage biogeochemistry concepts related to gold cycling against pancreatic cancer, considering mammalian organisms as drivers for gold nanoparticle biosynthesis. Sequestration of gold nanoparticles within tumors has been demonstrated as an effective strategy to enhance radiotherapy; however, the desmoplasia of pancreatic cancer impedes nanoparticle delivery. Our strategy overcomes this barrier by applying an atomic-scale agent, ionic gold, for intratumoral gold nanoparticle biosynthesis. Our comprehensive studies showed the cancer-specific synthesis of gold nanoparticles from externally delivered gold ions in vitro and in a murine pancreatic cancer model in vivo; a substantial colocalization of gold nanoparticles (GNPs) with cancer cell nuclei in vitro and in vivo; a strong radiosensitization effect by the intracellularly synthesized GNPs; a uniform distribution of in situ synthesized GNPs throughout the tumor volume; a nearly 40-day total suppression of tumor growth in animal models of pancreatic cancer treated with a combination of gold ions and radiation that was also associated with a significantly higher median survival versus radiation alone (235 vs 102 days, respectively).
Collapse
Affiliation(s)
- Aaron
S. Schwartz-Duval
- Department
of Imaging Physics, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yuri Mackeyev
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center, Houston, Texas 77030, United States
| | - Iqbal Mahmud
- Department
of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Philip L. Lorenzi
- Department
of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Mihai Gagea
- Department
of Veterinary Medicine & Surgery, The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Sunil Krishnan
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center, Houston, Texas 77030, United States
| | - Konstantin V. Sokolov
- Department
of Imaging Physics, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| |
Collapse
|
8
|
Pradyuth KS, Salunkhe SA, Singh AK, Chitkara D, Mittal A. Belinostat loaded lipid-polymer hybrid nanoparticulate delivery system for breast cancer: improved pharmacokinetics and biodistribution in a tumor model. J Mater Chem B 2023; 11:10859-10872. [PMID: 37938124 DOI: 10.1039/d3tb01317k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Despite various treatment modalities for breast cancer, it still persists as one of the most diagnosed types of cancer in females. The recent investigations in the epigenetics of breast cancer reveal several aberrations in the expression levels of various HDAC enzymes. Henceforth, the present work entails the formulation and characterization of a lipid polymer-based hybrid nanoparticulate (LPN) system for delivery of an epigenetic modulator drug, Belinostat, for its clinical application in breast cancer. The size of Belinostat nanoparticles prepared using a modified hot homogenization method was found to be 166.6 ± 19.95 nm with an encapsulation efficiency of 94.5 ± 5.1%. In vitro characterization for cytotoxicity, cellular uptake, and protein expression in two different breast cancer cells, 4T1 and MCF 7, revealed the superiority of the formulation in comparison with the free drug in MCF 7 cells. Subsequently, the behaviour of the formulation in in vivo settings of healthy and breast cancer xenograft bearing animals was analyzed using pharmacokinetic and biodistribution studies. The results revealed that the formulation demonstrated multi-fold improvement in the pharmacokinetic parameters in tumor bearing animals when compared with the free drug while no difference in pharmacokinetic behaviour was observed in healthy animals indicating the altered biodistribution and specificity of the formulation in breast tumor. This was confirmed by the biodistribution studies exhibiting 20-fold improved uptake and retention of the nanoparticulate formulation in tumor tissues of the animal model at the end of 4 h. Thus, the developed LPN system holds potential to act as a novel drug delivery system for Belinostat with several advantages over the free drug.
Collapse
Affiliation(s)
- Kommera Sai Pradyuth
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Shubham A Salunkhe
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Arihant Kumar Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
9
|
Pan Q, Fan X, Xie L, Wu D, Liu R, Gao W, Luo K, He B, Pu Y. Nano-enabled colorectal cancer therapy. J Control Release 2023; 362:548-564. [PMID: 37683732 DOI: 10.1016/j.jconrel.2023.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Colorectal cancer (CRC), one of the most common and deadliest diseases worldwide, poses a great health threat and social burden. The clinical treatments of CRC encompassing surgery, chemotherapy, and radiotherapy are challenged with toxicity, therapy resistance, and recurrence. In the past two decades, targeted therapy and immunotherapy have greatly improved the therapeutic benefits of CRC patients but they still suffer from drug resistance and low response rates. Very recently, gut microbiota regulation has exhibited a great potential in preventing and treating CRC, as well as in modulating the efficacy and toxicity of chemotherapy and immunotherapy. In this review, we provide a cutting-edge summary of nanomedicine-based treatment in colorectal cancer, highlighting the recent progress of oral and systemic tumor-targeting and/or tumor-activatable drug delivery systems as well as novel therapeutic strategies against CRC, including nano-sensitizing immunotherapy, anti-inflammation, gut microbiota modulation therapy, etc. Finally, the recent endeavors to address therapy resistance, metastasis, and recurrence in CRC were discussed. We hope this review could offer insight into the design and development of nanomedicines for CRC and beyond.
Collapse
Affiliation(s)
- Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Li Xie
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Di Wu
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rong Liu
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China.
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
10
|
Michlewska S, Wójkowska D, Watala C, Skiba E, Ortega P, de la Mata FJ, Bryszewska M, Ionov M. Ruthenium metallodendrimer against triple-negative breast cancer in mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102703. [PMID: 37591367 DOI: 10.1016/j.nano.2023.102703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/11/2023] [Accepted: 08/05/2023] [Indexed: 08/19/2023]
Abstract
Carbosilane metallodendrimers, based on the arene Ru(II) complex (CRD13) and integrated to imino-pyridine surface groups have been investigated as an anticancer agent in a mouse model with triple-negative breast cancer. The dendrimer entered into the cells efficiently, and exhibited selective toxicity for 4T1 cells. In vivo investigations proved that a local injection of CRD13 caused a reduction of tumour mass and was non-toxic. ICP analyses indicated that Ru(II) accumulated in all tested tissues with a greater content detected in the tumour.
Collapse
Affiliation(s)
- Sylwia Michlewska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Poland; Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland.
| | - Dagmara Wójkowska
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Poland
| | - Cezary Watala
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Poland
| | - Elżbieta Skiba
- Institute of General and Ecological Chemistry, Lodz University of Technology, Poland
| | - Paula Ortega
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain; Instituto de Investigación Sanitaria Ramón y Cajal, IRYCIS, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Francisco Javier de la Mata
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain; Instituto de Investigación Sanitaria Ramón y Cajal, IRYCIS, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| |
Collapse
|
11
|
Wang Q, Yang H, Liu Y, Zhou Z, Zhang X, Sang M, Xu F, Song L, Xia T, Zhang Y, Wei J, Zhang X, Ding Q. Awakening Allies for Breaking Microenvironment Barriers: NIR-II Guided Orthogonal Activation of Tumor-Infiltrating Mast Cells for Efficient Nano-Drug Delivery. Adv Healthc Mater 2023; 12:e2300420. [PMID: 37141500 DOI: 10.1002/adhm.202300420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/29/2023] [Indexed: 05/06/2023]
Abstract
Mast cells (MCs), powerful immune cells that heavily infiltrate cancer cells, play a crucial role in tumor formation. Activated MCs can release histamine and a family of proteases through degranulation effects, concurrently achieving endothelial junction weakening and stromal degradation of the tumor microenvironment, thereby clearing the obstacles for nano-drug infiltration. To achieve precise activation of tumor-infiltrating MCs, orthogonally excited rare earth nanoparticles (ORENP), with two channels, are introduced for the controllable stimulating drugs release wrapped in "photocut tape". The ORENP can emit near-infrared II (NIR-II) for image tracing for tumor localization in Channel 1 (808/NIR-II) and allows energy upconversion to emit ultraviolet (UV) light for releasing drugs for MCs stimulation in Channel 2 (980/UV). Finally, the combined use of chemical and cellular tools enables clinical nano-drugs to achieve a significant increase in tumor infiltration, thereby enhancing the efficacy of nano-chemotherapy.
Collapse
Affiliation(s)
- Qingyuan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Haiyan Yang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yuan Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhaoxi Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xu Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Mingyi Sang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Feng Xu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Lebin Song
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yue Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jifu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
- Department of Clinical Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing, 211103, China
| | - Xiaobo Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qiang Ding
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| |
Collapse
|
12
|
Mai W, Liu Q, Li J, Zheng M, Yan F, Liu H, Lei Y, Xu J, Xu J. Comprehensive analysis of the oncogenic and immunological role of FAP and identification of the ceRNA network in human cancers. Aging (Albany NY) 2023; 15:3738-3758. [PMID: 37166418 PMCID: PMC10449273 DOI: 10.18632/aging.204707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/22/2023] [Indexed: 05/12/2023]
Abstract
Fibroblast activation protein-alpha (FAP) is a transmembrane serine protease involving in tissue remodeling. Previous studies report that FAP is highly expressed in certain tumors and participated in oncogenesis. However, there is still lack of systematic and in-depth analysis of FAP based on clinical big data. Here, we comprehensively map the FAP expression profile, prognostic outcome, genetic alteration, immune infiltration across over 30 types of human cancers through multiple datasets including TCGA, CPTAC, and cBioPortal. We find that FAP is up-regulated in most cancer types, and increased FAP expression is associated with advanced pathological stages or poor prognosis in several cancers. Furthermore, FAP is significantly correlated with the infiltration of cancer-associated fibroblasts, macrophages, myeloid dendritic cells, as well as endothelia cells. Immunosuppressive checkpoint proteins or cytokines expression, microsatellite instability and tumor mutational burden analysis also indicate the regulation role of FAP in tumor progression. Gene enrichment analysis demonstrates that ECM-receptor interaction as well as extracellular matrix and structure process are linked to the potential mechanism of FAP in tumor pathogenesis. The ceRNA network is also constructed and identified the involvement of LINC00707/hsa-miR-30e-5p/FAP, LINC02535/hsa-miR-30e-5p/FAP, LINC02535/hsa-miR-30d-5p/FAP, as well as AC026356.1/hsa-miR-30d-5p/FAP axis in tumor progression. In conclusion, our study offers new insights into the oncogenic and immunological role of FAP from a pan-cancer perspective, providing new clues for developing novel targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Weiqian Mai
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Qingyou Liu
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Jiasheng Li
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Mincheng Zheng
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Fuman Yan
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Hui Liu
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Yuhe Lei
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Jinwen Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Jiean Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| |
Collapse
|
13
|
Geng S, Xiang T, Zhang Y, Guo P, Zhang H, Zhang Z, Gu M, Zhang K, Song H, Shi J, Liu J. Safe engineering of cancer-associated fibroblasts enhances checkpoint blockade immunotherapy. J Control Release 2023; 356:272-287. [PMID: 36870541 DOI: 10.1016/j.jconrel.2023.02.041] [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: 11/23/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Abundant cancer-associated fibroblasts (CAFs) in highly fibrotic breast cancer constitute an immunosuppressive barrier for T cell activity and are closely related to the failure of immune checkpoint blockade therapy (ICB). Inspired by the similar antigen-processing capacity of CAFs to professional antigen-presenting cells (APCs), a "turning foes to friends" strategy is proposed by in situ engineering immune-suppressed CAFs into immune-activated APCs for improving response rates of ICB. To achieve safe and specific CAFs engineering in vivo, a thermochromic spatiotemporal photo-controlled gene expression nanosystem was developed by self-assembly of molten eutectic mixture, chitosan andfusion plasmid. After photoactivatable gene expression, CAFs could be engineered as APCs via co-stimulatory molecule (CD86) expression, which effectively induced activation and proliferation of antigen-specific CD8 + T cells. Meanwhile, engineered CAFs could also secrete PD-L1 trap protein in situ for ICB, avoiding potential autoimmune-like disorders caused by "off-target" effects of clinically applied PD-L1 antibody. The study demonstrated that the designed nanosystem could efficiently engineer CAFs, significantly enhance the percentages of CD8+ T cells (4-folds), result in about 85% tumor inhibition rate and 83.3% survival rate at 60 days in highly fibrotic breast cancer, further inducing long-term immune memory effects and effectively inhibiting lung metastasis.
Collapse
Affiliation(s)
- Shizhen Geng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tingting Xiang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yunya Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pengke Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Mengchao Gu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Haiwei Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore 138673, Singapore.
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| |
Collapse
|
14
|
Zhang H, Chen L, Zhao Y, Luo N, Shi J, Xu S, Ma L, Wang M, Gu M, Mu C, Xiong Y. Relaxin-encapsulated polymeric metformin nanoparticles remodel tumor immune microenvironment by reducing CAFs for efficient triple-negative breast cancer immunotherapy. Asian J Pharm Sci 2023; 18:100796. [PMID: 37008735 PMCID: PMC10064789 DOI: 10.1016/j.ajps.2023.100796] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/14/2023] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are one of the most abundant stromal cells in the tumor microenvironment which mediate desmoplastic response and are the primary driver for an immunosuppressive microenvironment, leading to the failure of triple-negative breast cancer (TNBC) immunotherapy. Therefore, depleting CAFs may enhance the effect of immunotherapy (such as PD-L1 antibody). Relaxin (RLN) has been demonstrated to significantly improve transforming growth factor-β (TGF-β) induced CAFs activation and tumor immunosuppressive microenvironment. However, the short half-life and systemic vasodilation of RLN limit its in vivo efficacy. Here, plasmid encoding relaxin (pRLN) to locally express RLN was delivered with a new positively charged polymer named polymeric metformin (PolyMet), which could increase gene transfer efficiency significantly and have low toxicity that have been certified by our lab before. In order to improve the stability of pRLN in vivo, this complex was further formed lipid poly-γ-glutamic acid (PGA)/PolyMet-pRLN nanoparticle (LPPR). The particle size of LPPR was 205.5 ± 2.9 nm, and the zeta potential was +55.4 ± 1.6 mV. LPPR displayed excellent tumor penetrating efficacy and weaken proliferation of CAFs in 4T1luc/CAFs tumor spheres in vitro. In vivo, it could reverse aberrantly activated CAFs by decreasing the expression of profibrogenic cytokine and remove the physical barrier to reshape the tumor stromal microenvironment, which enabled a 2.2-fold increase in cytotoxic T cell infiltration within the tumor and a decrease in immunosuppressive cells infiltration. Thus, LPPR was observed retarded tumor growth by itself in the 4T1 tumor bearing-mouse, and the reshaped immune microenvironment further led to facilitate antitumor effect when it combined with PD-L1 antibody (aPD-L1). Altogether, this study presented a novel therapeutic approach against tumor stroma using LPPR to achieve a combination regimen with immune checkpoint blockade therapy against the desmoplastic TNBC model.
Collapse
Affiliation(s)
- Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liying Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yue Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ningchao Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shujun Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lisha Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Menglin Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Mancang Gu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chaofeng Mu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Corresponding author at: School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
15
|
Liu X, Zhou J, Wu H, Chen S, Zhang L, Tang W, Duan L, Wang Y, McCabe E, Hu M, Yu Z, Liu H, Choi CHJ, Sung JJY, Huang L, Liu R, Cheng ASL. Fibrotic immune microenvironment remodeling mediates superior anti-tumor efficacy of a nano-PD-L1 trap in hepatocellular carcinoma. Mol Ther 2023; 31:119-133. [PMID: 36146933 PMCID: PMC9840184 DOI: 10.1016/j.ymthe.2022.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/20/2022] [Accepted: 09/19/2022] [Indexed: 01/27/2023] Open
Abstract
The local microenvironment where tumors develop can shape cancer progression and therapeutic outcome. Emerging evidence demonstrate that the efficacy of immune-checkpoint blockade (ICB) is undermined by fibrotic tumor microenvironment (TME). The majority of hepatocellular carcinoma (HCC) develops in liver fibrosis, in which the stromal and immune components may form a barricade against immunotherapy. Here, we report that nanodelivery of a programmed death-ligand 1 (PD-L1) trap gene exerts superior efficacy in treating fibrosis-associated HCC when compared with the conventional monoclonal antibody (mAb). In two fibrosis-associated HCC models induced by carbon tetrachloride and a high-fat, high-carbohydrate diet, the PD-L1 trap induced significantly larger tumor regression than mAb with no evidence of toxicity. Mechanistic studies revealed that PD-L1 trap, but not mAb, consistently reduced the M2 macrophage proportion in the fibrotic liver microenvironment and promoted cytotoxic interferon gamma (IFNγ)+tumor necrosis factor α (TNF-α)+CD8+T cell infiltration to the tumor. Moreover, PD-L1 trap treatment was associated with decreased tumor-infiltrating polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) accumulation, resulting in an inflamed TME with a high cytotoxic CD8+T cell/PMN-MDSC ratio conductive to anti-tumor immune response. Single-cell RNA sequencing analysis of two clinical cohorts demonstrated preferential PD-L1 expression in M2 macrophages in the fibrotic liver, thus supporting the translational potential of nano-PD-L1 trap for fibrotic HCC treatment.
Collapse
Affiliation(s)
- Xiaoyu Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Jingying Zhou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Haoran Wu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Shufen Chen
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Lingyun Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wenshu Tang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Liang Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Ying Wang
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eleanor McCabe
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mengying Hu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhuo Yu
- Department of Liver Disease, Shuguang Hospital, affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hanzhuang Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Chung Hang Jonathan Choi
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Joseph Jao-Yiu Sung
- Lee Kong Chian School of Medicine, Nanyang Technological University, 639798 Singapore, Singapore; State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rihe Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Alfred Sze-Lok Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
16
|
Dana P, Thumrongsiri N, Tanyapanyachon P, Chonniyom W, Punnakitikashem P, Saengkrit N. Resveratrol Loaded Liposomes Disrupt Cancer Associated Fibroblast Communications within the Tumor Microenvironment to Inhibit Colorectal Cancer Aggressiveness. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 13:107. [PMID: 36616017 PMCID: PMC9824711 DOI: 10.3390/nano13010107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Colorectal cancer (CRC) is a cancer-associated fibroblast, CAF-rich tumor. CAF promotes cancer cell proliferation, metastasis, drug resistance via secretes soluble factors, and extracellular matrices which leads to dense stroma, a major barrier for drug delivery. Resveratrol (RES) is a polyphenolic compound, has several pharmacologic functions including anti-inflammation and anticancer effects. Considering tumor microenvironment of CRC, resveratrol-loaded liposome (L-RES) was synthesized and employed to inhibit CAF functions. The L-RES was synthesized by thin-film hydration method. The cytotoxicity of L-RES was evaluated using MTT assay. Effect of L-RES treated CAF on tumor spheroid growth was performed. Cell invasion was determined using spheroid invasion assay. The effect of L-RES on 5-fluorouracil (5-FU) sensitivity of CRC cells was determined in co-cultured tumor spheroids. Subtoxic dose of L-RES was selected to study possible inhibiting CAF functions. Decreased CAF markers, α-SMA and IL-6 levels, were observed in L-RES treated activated fibroblast. Interestingly, the activated fibroblast promoted invasive ability and drug resistance of CRC cells in co-culture condition of both 2D and 3D cultures and was attenuated by L-RES treatment in the activated fibroblast. Therefore, L-RES provides a promising drug delivery strategy for CRC treatment by disrupting the crosstalk between CRC cells and CAF.
Collapse
Affiliation(s)
- Paweena Dana
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Nutthanit Thumrongsiri
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Prattana Tanyapanyachon
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Walailuk Chonniyom
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand
- Research Network NANOTEC-Mahidol University in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand
| | - Nattika Saengkrit
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| |
Collapse
|
17
|
Zhang Q, Wang X, Kuang G, Zhao Y. Pt(IV) prodrug initiated microparticles from microfluidics for tumor chemo-, photothermal and photodynamic combination therapy. Bioact Mater 2022; 24:185-196. [PMID: 36606251 PMCID: PMC9804016 DOI: 10.1016/j.bioactmat.2022.12.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Multimodal treatment modalities hold great potential for cancer therapy, thus current efforts are focusing on the development of more effective and practical synergistic therapeutic platforms. Herein, we present a novel trans, trans,trans-[Pt(N3)2(OH)2(py)2] (Pt(IV)) prodrug-initiated hydrogel microparticles (MICG-Pt) with indocyanine green (ICG) encapsulation by microfluidics for efficiently synergistic chemo-, photothermal (PTT) and photodynamic therapy (PDT). The employed Pt(IV) could not only serves as an initiator to generate azidyl radical (N3 •) for photo-polymerization of methacrylate gelatin (GelMA) matrix, but also be reduced to high cytotoxic platinum(II) (Pt(II)) species for tumor chemotherapy. The laden ICG with highly photothermal heating ability and intrinsic reactive oxygen species (ROS) productivity endows the MICG-Pt with effective PTT/PDT performances upon near-infrared (NIR) light irradiation. In addition, benefiting from the production of oxygen during the photo-activation process of Pt(IV), the PDT efficacy of ICG-laden MICG-Pt could be further enhanced. Based on these advantages, we have demonstrated that the MICG-Pt could significantly eliminate cancer cells in vitro, and remarkably suppressed the tumor growth in vivo via synergistic chemotherapy, PTT, and PDT. These results indicate that such Pt(IV)-initiated hydrogel microparticles are ideal candidates of multimodal treatment platforms, holding great prospects for cancer therapy.
Collapse
Affiliation(s)
- Qingfei Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xiaocheng Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Gaizhen Kuang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China,Corresponding author. Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
18
|
Peng H, He X, Wang Q. Targeted drug delivery system for ovarian cancer microenvironment: Improving the effects of immunotherapy. Front Immunol 2022; 13:1035997. [PMID: 36405688 PMCID: PMC9670735 DOI: 10.3389/fimmu.2022.1035997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Immunotherapies have shown modest benefits in the current clinical trials for ovarian cancer. The tumor microenvironment (TME) in an immunosuppressive phenotype contributes to this “failure” of immunotherapy in ovarian cancer. Many stromal cell types in the TME (e.g., tumor-associated macrophages and fibroblasts) have been identified as having plasticity in pro- and antitumor activities and are responsible for suppressing the antitumor immune response. Thus, the TME is an extremely valuable target for adjuvant interventions to improve the effects of immunotherapy. The current strategies targeting the TME include: 1) eliminating immunosuppressive cells or transforming them into immunostimulatory phenotypes and 2) inhibiting their immunosuppressive or pro-tumor production. Most of the effective agents used in the above strategies are genetic materials (e.g., cDNA, mRNA, or miRNA), proteins, or other small molecules (e.g., peptides), which are limited in their target and instability. Various formulations of drug delivery system (DDS) have been designed to realize the controlled release and targeting delivery of these agents to the tumor sites. Nanoparticles and liposomes are the most frequently exploited materials. Based on current evidence from preclinical and clinical studies, the future of the DDS is promising in cancer immunotherapy since the combination of agents with a DDS has shown increased efficacy and decreased toxicities compared with free agents. In the future, more efforts are needed to further identify the hallmarks and biomarkers in the ovarian TME, which is crucial for the development of more effective, safe, and personalized DDSs.
Collapse
|
19
|
Han S, Wu J. Three-dimensional (3D) scaffolds as powerful weapons for tumor immunotherapy. Bioact Mater 2022; 17:300-319. [PMID: 35386452 PMCID: PMC8965033 DOI: 10.1016/j.bioactmat.2022.01.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Though increasing understanding and remarkable clinical successes have been made, enormous challenges remain to be solved in the field of cancer immunotherapy. In this context, biomaterial-based immunomodulatory strategies are being developed to boost antitumor immunity. For the local immunotherapy, macroscale biomaterial scaffolds with 3D network structures show great superiority in the following aspects: facilitating the encapsulation, localized delivery, and controlled release of immunotherapeutic agents and even immunocytes for more efficient immunomodulation. The concentrating immunomodulation in situ could minimize systemic toxicities, but still exert abscopal effects to harness the power of overall anticancer immune response for eradicating malignancy. To promote such promising immunotherapies, the design requirements of macroscale 3D scaffolds should comprehensively consider their physicochemical and biological properties, such as porosity, stiffness, surface modification, cargo release kinetics, biocompatibility, biodegradability, and delivery modes. To date, increasing studies have focused on the relationships between these parameters and the biosystems which will guide/assist the 3D biomaterial scaffolds to achieve the desired immunotherapeutic outcomes. In this review, by highlighting some recent achievements, we summarized the latest advances in the development of various 3D scaffolds as niches for cancer immunotherapy. We also discussed opportunities, challenges, current trends, and future perspectives in 3D macroscale biomaterial scaffold-assisted local treatment strategies. More importantly, this review put more efforts to illustrate how the 3D biomaterial systems affect to modulate antitumor immune activities, where we discussed how significant the roles and behaviours of 3D macroscale scaffolds towards in situ cancer immunotherapy in order to direct the design of 3D immunotherapeutic.
Collapse
Affiliation(s)
- Shuyan Han
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, 518057, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, 518057, China
| |
Collapse
|
20
|
Wu J, Wang F, Dong J, Zhang S, Li N, Zhao H, Liu X, Gao Z, Zhang B, Tian G. Therapeutic Response of Multifunctional Lipid and Micelle Formulation in Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:45110-45123. [PMID: 36167351 DOI: 10.1021/acsami.2c10446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Hepatic stellate cells (HSCs), as an important part of the tumor microenvironment (TME), could be activated by tumor cells as cancer-associated fibroblasts (CAFs), thereby promoting the production of extracellular matrix (ECM) and favoring the development of tumors. Therefore, blocking the "CAFs-ECM" axis is a promising pathway to improve antitumor efficacy. Based on this, we developed a multifunctional nanosized delivery system composed of hyaluronic acid-modified pH-sensitive liposomes (CTHLs) and glycyrrheic acid-modified nanomicelles (DGNs), which combines the advantages of targeted delivery, pH-sensitivity, and deep drug penetration. To mimic actual TME, a novel HSCs+BEL-7402 cocultured cell model and a m-HSCs+H22 coimplanted mice model were established. As expected, CTHLs and DGNs could target CAFs and tumor cells, respectively, and promote the drug penetration and retention in tumor regions. Notably, CTHLs+DGNs not only exhibited a superior antitumor effect in three-level tumor-bearing mice but also presented excellent antimetastasis efficiency in lung-metastatic mice. The antitumor mechanism revealed that the lipid&micelle mixed formulations effectively inhibited the activation of CAFs, reduced the deposition of ECM, and reversed the epithelial-mesenchymal transition (EMT) of tumor cells. In brief, the nanosized delivery system composed of CTHLs and DGNs could effectively improve the therapeutic effect of liver cancer by blocking the "CAFs-ECM" axis, which has a good clinical application prospect.
Collapse
Affiliation(s)
- Jingliang Wu
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
| | - Fangqing Wang
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, P.R. China
| | - Jinping Dong
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
| | - Suqiu Zhang
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
| | - Na Li
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
| | - Huifang Zhao
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
| | - Xuemin Liu
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, P.R. China
| | - Zhiqin Gao
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, P.R. China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, P.R. China
| | - Guixiang Tian
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, P.R. China
| |
Collapse
|
21
|
He J, Liu D, Liu M, Tang R, Zhang D. Characterizing the role of SLC3A2 in the molecular landscape and immune microenvironment across human tumors. Front Mol Biosci 2022; 9:961410. [PMID: 35992269 PMCID: PMC9388758 DOI: 10.3389/fmolb.2022.961410] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Inducing ferroptosis in human tumors has become a potential strategy to improve the prognosis of patients, even in those with chemotherapeutic resistance. The xCT complex is a major target for ferroptosis induction, constituted by SLC7A11 and SLC3A2. The role of SLC7A11 in cancer has been widely studied in recent years. However, related research studies for its partner SLC3A2 are still rare. Methods: Bulk transcriptome, single-cell sequencing, and immunohistochemical staining were analyzed to explore the expression distribution of SLC3A2. Clinical outcomes were referred to uncover the relationship between SLC3A2 expression and patients’ prognosis. Immune cell infiltration was estimated by multiple deconvolution algorithms. The effect of SLC3A2 on the proliferation and drug resistance of cancer cell lines was evaluated by DEPMAP. Results: Upregulated SLC3A2 may have an adverse effect on the survival of multiple cancers such as lower-grade glioma and acute myeloid leukemia. SLC3A2 expression is indispensable for multiple cell lines’ proliferation, especially for ESO51 (a cell line for esophageal cancer). In addition, SLC3A2 expression level was related to the remodeling of the immune microenvironment in cancers and some immune checkpoints such as PD-1 and PD-L1, which were potential therapeutic targets in many distinct cancers. Conclusion: Our study systematically elucidated the role of SLC3A2 in the survival of cancer patients and the potential immunotherapeutic response. Few molecular mechanisms by which SLC3A2 regulates anti-tumor immunity have been clarified in the present study, which is the main limitation. Future research into the biological mechanism could further help with targeted treatment for cancer patients.
Collapse
Affiliation(s)
- Jiajun He
- Minhang Hospital, Fudan University, Shanghai, China
| | - Dong Liu
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mei Liu
- Minhang Hospital, Fudan University, Shanghai, China
| | - Rong Tang
- Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Rong Tang, ; Dongqing Zhang,
| | - Dongqing Zhang
- Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Rong Tang, ; Dongqing Zhang,
| |
Collapse
|
22
|
Boyd LNC, Andini KD, Peters GJ, Kazemier G, Giovannetti E. Heterogeneity and plasticity of cancer-associated fibroblasts in the pancreatic tumor microenvironment. Semin Cancer Biol 2022; 82:184-196. [PMID: 33737108 DOI: 10.1016/j.semcancer.2021.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/17/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a notably poor prognosis, in urgent need of improved treatment strategies. The desmoplastic PDAC tumor microenvironment (TME), marked by a high concentration of cancer-associated-fibroblasts (CAFs), is a dynamic part of PDAC pathophysiology which occasions a variety of effects throughout the course of pancreatic tumorigenesis and disease evolution. A better understanding of the desmoplastic TME and CAF biology in particular, should provide new opportunities for improving therapeutics. That CAFs have a tumor-supportive role in oncogenesis is well known, yet research evidence has shown that CAFs also have tumor-repressive functions. In this review, we seek to clarify the intriguing heterogeneity and plasticity of CAFs and their ambivalent role in PDAC tumorigenesis and progression. Additionally, we provide recommendations to advance the implementation of CAF-directed PDAC care. An improved understanding of CAFs' origins, spatial location, functional diversity, and marker determination, as well as CAF behavior during the course of PDAC progression and metastasis will provide essential knowledge for the future improvement of therapeutic strategies for patients suffering from PDAC.
Collapse
Affiliation(s)
- Lenka N C Boyd
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University, De Boelelaan 1118, 1081 HZ, Postbus 7057, 1007 MB, Amsterdam, the Netherlands; Department of Medical Oncology, Lab of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, De Boelelaan 1118, 1081 HZ, Postbus 7057, 1007 MB, Amsterdam, the Netherlands.
| | - Katarina D Andini
- Department of Medical Oncology, Lab of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, De Boelelaan 1118, 1081 HZ, Postbus 7057, 1007 MB, Amsterdam, the Netherlands.
| | - Godefridus J Peters
- Department of Medical Oncology, Lab of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, De Boelelaan 1118, 1081 HZ, Postbus 7057, 1007 MB, Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Marii Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland.
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University, De Boelelaan 1118, 1081 HZ, Postbus 7057, 1007 MB, Amsterdam, the Netherlands.
| | - Elisa Giovannetti
- Department of Medical Oncology, Lab of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, De Boelelaan 1118, 1081 HZ, Postbus 7057, 1007 MB, Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Via Ferruccio Giovannini, 13, 56017, San Giuliano Terme PI, Pisa, Italy.
| |
Collapse
|
23
|
Li Y, Wu J, Lu Q, Liu X, Wen J, Qi X, Liu J, Lian B, Zhang B, Sun H, Tian G. GA&HA-Modified Liposomes for Co-Delivery of Aprepitant and Curcumin to Inhibit Drug-Resistance and Metastasis of Hepatocellular Carcinoma. Int J Nanomedicine 2022; 17:2559-2575. [PMID: 35698562 PMCID: PMC9188407 DOI: 10.2147/ijn.s366180] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Background Tumor microenvironment (TME) plays a vital role in the development of hepatocellular carcinoma (HCC). Mounting evidence indicates that peripheral nerves could induce a shift from quiescent hepatic stellate cells (HSCs) to cancer-associated fibroblasts (CAFs) by secreting substance P (SP). The anti-tumor strategy by targeting “SP-HSCs-HCC” axis might be an effective therapy to inhibit tumor growth and metastasis. Objective In this study, we prepared novel liposomes (CUR-APR/HA&GA-LPs) modified with hyaluronic acid (HA) and glycyrrhetinic acid (GA) for co-delivery aprepitant (APR) and curcumin (CUR), in which APR was chosen to inhibit the activation of HSCs by blocking SP/neurokinin-1 receptor (NK-1R), and CUR was used to induce apoptosis of tumor cells. Results To mimic the TME, we established “SP+HSCs+HCC” co-cultured cell model in vitro. The results showed that CUR-APR/HA&GA-LPs could be taken up by CAFs and HCC simultaneously, and inhibit tumor cell migration. Meanwhile, the “SP+m-HSCs+HCC” co-implanted mice model was established to evaluate the anti-tumor effect in vivo. The results showed that CUR-APR/HA&GA-LPs could inhibit tumor proliferation and metastasis, and reduce extracellular matrix (ECM) deposition and tumor angiogenesis, indicating a superior anti-HCC effect. Conclusion Overall, the combination therapy based on HA&GA-LPs could be a potential nano-sized formulation for anti-HCC therapy.
Collapse
Affiliation(s)
- Yanying Li
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, People's Republic of China.,School of Nursing, Weifang University of Science and Technology, Weifang, 262700, People's Republic of China
| | - Jingliang Wu
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, People's Republic of China
| | - Qiao Lu
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Xuemin Liu
- School of Nursing, Weifang University of Science and Technology, Weifang, 262700, People's Republic of China
| | - Jiaxuan Wen
- School of Nursing, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Xiaohui Qi
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Jianhao Liu
- School of Pharmacy, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Bo Lian
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Hengyi Sun
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Guixiang Tian
- School of Life Science and Technology, Weifang Medical University, Weifang, 261053, People's Republic of China
| |
Collapse
|
24
|
Chang J, Mo L, Song J, Wang X, Liu H, Meng C, Wu Y. A pH-responsive mesoporous silica nanoparticle-based drug delivery system for targeted breast cancer therapy. J Mater Chem B 2022; 10:3375-3385. [PMID: 35388835 DOI: 10.1039/d1tb02828f] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In order to make the drug specifically aggregate at the tumor site, we had developed a targeted drug delivery system based on pH responsive mesoporous silica nanoparticles. Mesoporous silica nanoparticles (MSN-COOH) were prepared and doxorubicin (DOX) was loaded into the pores of MSN-COOH, and then polyethyleneimine (PEI) and anisamide (AA) were modified on the surface of mesoporous silica, named DOX@MSN-PEI-AA(DMPA). DMPA specifically entered tumor cells through AA-mediated receptor endocytosis; PEI dissociated from the surface of the MSN in the acidic environment of cellular lysosomes/endosomes due to protonation of PEI, resulting in steady release of the encapsulated DOX from the pores of MSN in the cytoplasm of the target cells. In vitro and in vivo anti-tumor experiments and hemolytic experiments indicated that DMPA can accurately target breast cancer cells and show excellent safety at the same time, showing great potential for tumor therapy.
Collapse
Affiliation(s)
- Jie Chang
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, China.
| | - Liufang Mo
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, China.
| | - Jinfeng Song
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, China.
| | - Xiaochen Wang
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, China.
| | - Hanhan Liu
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, China.
| | - Chenchen Meng
- Kaifeng Central Hospital, Kaifeng Henan, 475004, China
| | - Yijun Wu
- College of Pharmacy of Henan University, Kaifeng Henan, 475004, China.
| |
Collapse
|
25
|
Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Semin Cancer Biol 2022; 86:624-644. [DOI: 10.1016/j.semcancer.2022.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022]
|
26
|
Strategies targeting tumor immune and stromal microenvironment and their clinical relevance. Adv Drug Deliv Rev 2022; 183:114137. [PMID: 35143893 DOI: 10.1016/j.addr.2022.114137] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 12/13/2022]
Abstract
The critical role of tumor microenvironment (TME) in tumor initiation and development has been well-recognized after more than a century of studies. Numerous therapeutic approaches targeting TME are rapidly developed including those leveraging nanotechnology, which have been further accelerated since the emergence of immune checkpoint blockade therapies in the past decade. While there are many reviews focusing on TME remodeling therapies via drug delivery and engineering strategies in animal models, state-of-the-art evaluation of clinical development states of TME-targeted therapeutics is rarely found. Here, we illustrate opportunities for integrating nano-delivery system for the development of TME-specific therapeutic regimen, followed by a comprehensive summary of the most up to date approved or clinically evaluated therapeutics targeting cellular and extracellular components within tumor immune and stromal microenvironment, including small molecule and monoclonal antibody drugs as well as nanomedicines. In the end, we also discuss challenges and possible solutions for clinical translation of TME-targeted nanomedicines.
Collapse
|
27
|
Li Y, Chen Z, Gu L, Duan Z, Pan D, Xu Z, Gong Q, Li Y, Zhu H, Luo K. Anticancer nanomedicines harnessing tumor microenvironmental components. Expert Opin Drug Deliv 2022; 19:337-354. [PMID: 35244503 DOI: 10.1080/17425247.2022.2050211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Small-molecular drugs are extensively used in cancer therapy, while they have issues of nonspecific distribution and consequent side effects. Nanomedicines that incorporate chemotherapeutic drugs have been developed to enhance the therapeutic efficacy of these drugs and reduce their side effects. One of the promising strategies is to prepare nanomedicines by harnessing the unique tumor microenvironment (TME). AREAS COVERED The TME contains numerous cell types that specifically express specific antibodies on the surface including tumor vascular endothelial cells, tumor-associated adipocytes, tumor-associated fibroblasts, tumor-associated immune cells and cancer stem cells. The physicochemical environment is characterized with a low pH, hypoxia, and a high redox potential resulting from tumor-specific metabolism. The intelligent nanomedicines can be categorized into two groups: the first group which is rapidly responsive to extracellular chemical/biological factors in the TME and the second one which actively and/or specifically targets cellular components in the TME. EXPERT OPINION In this paper, we review recent progress of nanomedicines by harnessing the TME and illustrate the principles and advantages of different strategies for designing nanomedicines, which are of great significance for exploring novel nanomedicines or translating current nanomedicines into clinical practice. We will discuss the challenges and prospects of preparing nanomedicines to utilize or alter the TME for achieving effective, safe anticancer treatment.
Collapse
Affiliation(s)
- Yinggang Li
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhonglan Chen
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Gu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyu Duan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dayi Pan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuping Xu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Youping Li
- Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
28
|
Ni K, Xu Z, Culbert A, Luo T, Guo N, Yang K, Pearson E, Preusser B, Wu T, La Riviere P, Weichselbaum RR, Spiotto MT, Lin W. Synergistic checkpoint-blockade and radiotherapy–radiodynamic therapy via an immunomodulatory nanoscale metal–organic framework. Nat Biomed Eng 2022; 6:144-156. [DOI: 10.1038/s41551-022-00846-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 01/12/2022] [Indexed: 12/20/2022]
|
29
|
Vyas D, Patel M, Wairkar S. Strategies for active tumor targeting-an update. Eur J Pharmacol 2022; 915:174512. [PMID: 34555395 DOI: 10.1016/j.ejphar.2021.174512] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 01/26/2023]
Abstract
A complete cure for cancer is still the holy grail for scientists. The existing treatment of cancer is primarily focused on surgery, radiation and conventional chemotherapy. However, chemotherapeutic agents also affect healthy tissues or organs due to a lack of specificity. While passive targeting is studied for anticancer drugs focused on the enhanced permeability and retention effect, it failed to achieve drug accumulation at the tumor site and desired therapeutic efficacy. This review presents an outline of the current significant targets for active tumor drug delivery systems and provides insight into the direction of active tumor-targeting strategies. For this purpose, a systematic understanding of the physiological factors, tumor microenvironment and its components, overexpressed receptor and associated proteins are covered here. We focused on angiogenesis mediated targeting, receptor-mediated targeting and peptide targeting. This active targeting along with integration with nano delivery systems helps in achieving specific action, thus reducing the associated adverse effects to healthy tissues. Although the tumor-targeting methods and possibilities explored so far seem revolutionary in cancer treatment, in-depth clinical studies data is required for its commercial translation.
Collapse
Affiliation(s)
- Darshan Vyas
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Mital Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
30
|
Avagliano A, Arcucci A. Insights into Melanoma Fibroblast Populations and Therapeutic Strategy Perspectives: Friends or Foes? Curr Med Chem 2022; 29:6159-6168. [PMID: 35726413 DOI: 10.2174/0929867329666220620124138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022]
Abstract
Cutaneous melanoma (CM) is an aggressive and highly metastatic solid tumor associated with drug resistance. Before 2011, despite therapies based on cytokines or molecules inhibiting DNA synthesis, metastatic melanoma led to patient death within 18 months from diagnosis. However, recent studies on bidirectional interactions between melanoma cells and tumor microenvironment (TME) have had a significant impact on the development of new therapeutic strategies represented by targeted therapy and immunotherapy. In particular, the heterogeneous stromal fibroblast populations, including fibroblasts, fibroblast aggregates, myofibroblasts, and melanoma associated fibroblasts (MAFs), represent the most abundant cell population of TME and regulate cancer growth differently. Therefore, in this perspective article, we have highlighted the different impacts of fibroblast populations on cancer development and growth. In particular, we focused on the role of MAFs in sustaining melanoma cell survival, proliferation, migration and invasion, drug resistance, and immunoregulation. The important role of constitutively activated MAFs in promoting CM growth and immunoediting makes this cell type a promising target for cancer therapy.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
31
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
32
|
Tang J, Zhang X, Cheng L, Liu Y, Chen Y, Jiang Z, Liu J. Multiple stimuli-responsive nanosystem for potent, ROS-amplifying, chemo-sonodynamic antitumor therapy. Bioact Mater 2021; 15:355-371. [PMID: 35356815 PMCID: PMC8935089 DOI: 10.1016/j.bioactmat.2021.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
Although sonodynamic therapy (SDT) is a promising non-invasive tumor treatment strategy due to its safety, tissue penetration depth and low cost, the hypoxic tumor microenvironment limits its therapeutic effects. Herein, we have designed and developed an oxygen-independent, ROS-amplifying chemo-sonodynamic antitumor therapy based on novel pH/GSH/ROS triple-responsive PEG-PPMDT nanoparticles. The formulated artemether (ART)/Fe3O4-loaded PEG-PPMDT NPs can rapidly release drug under the synergistic effect of acidic endoplasmic pH and high intracellular GSH/ROS levels to inhibit cancer cell growth. Besides, the ROS level in the NPs-treated tumor cells is magnified by ART via interactions with both Fe2+ ions formed in situ at acidic pH and external ultrasound irradiation, which is not affected by hypoxia tumor microenvironment. Consequently, the enriched intracellular ROS level can cause direct necrosis of ROS-stressed tumor cells and further accelerate the drug release from the ROS-responsive PEG-PPMDT NPs, achieving an incredible antitumor potency. Specifically, upon the chemo-sonodynamic therapy by ART/Fe3O4-loaded PEG-PPMDT NPs, all xenotransplants of human hepatocellular carcinoma (HepG2) in nude mice shrank significantly, and 40% of the tumors were completely eliminated. Importantly, the Fe3O4 encapsulated in the NPs is an efficient MRI contrast agent and can be used to guide the therapeutic procedures. Further, biosafety analyses show that the PEG-PPMDT NPs possess minimal toxicity to main organs. Thus, our combined chemo-sonodynamic therapeutic method is promising for potent antitumor treatment by controlled release of drug and facile exogenous generation of abundant ROS at target tumor sites. pH/GSH/ROS triple-responsive PEG-PPMDT were synthesized by enzymatic polymerization. ART and Fe3O4 loaded PEG-PPMDT NPs processes SDT/CDT and MRI theranostic function. Intracellular ROS was magnified by Fe2+-ART interaction and ultrasound irradiation.
Collapse
Affiliation(s)
- JunJie Tang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Xiaoge Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Lili Cheng
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Yadong Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - You Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Zhaozhong Jiang
- Faculty of Arts and Sciences, Department of Biomedical Engineering, Integrated Science and Technology Center, Yale University, 600 West Campus Drive, West Haven, CT, 06516, United States
- Corresponding author.
| | - Jie Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- Corresponding author.
| |
Collapse
|
33
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
34
|
Zhang J, Hu K, Di L, Wang P, Liu Z, Zhang J, Yue P, Song W, Zhang J, Chen T, Wang Z, Zhang Y, Wang X, Zhan C, Cheng YC, Li X, Li Q, Fan JY, Shen Y, Han JY, Qiao H. Traditional herbal medicine and nanomedicine: Converging disciplines to improve therapeutic efficacy and human health. Adv Drug Deliv Rev 2021; 178:113964. [PMID: 34499982 DOI: 10.1016/j.addr.2021.113964] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Traditional herbal medicine (THM), an ancient science, is a gift from nature. For thousands of years, it has helped humans fight diseases and protect life, health, and reproduction. Nanomedicine, a newer discipline has evolved from exploitation of the unique nanoscale morphology and is widely used in diagnosis, imaging, drug delivery, and other biomedical fields. Although THM and nanomedicine differ greatly in time span and discipline dimensions, they are closely related and are even evolving toward integration and convergence. This review begins with the history and latest research progress of THM and nanomedicine, expounding their respective developmental trajectory. It then discusses the overlapping connectivity and relevance of the two fields, including nanoaggregates generated in herbal medicine decoctions, the application of nanotechnology in the delivery and treatment of natural active ingredients, and the influence of physiological regulatory capability of THM on the in vivo fate of nanoparticles. Finally, future development trends, challenges, and research directions are discussed.
Collapse
|
35
|
Li W, Little N, Park J, Foster CA, Chen J, Lu J. Tumor-Associated Fibroblast-Targeting Nanoparticles for Enhancing Solid Tumor Therapy: Progress and Challenges. Mol Pharm 2021; 18:2889-2905. [PMID: 34260250 DOI: 10.1021/acs.molpharmaceut.1c00455] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Even though nanoparticle drug delivery systems (nanoDDSs) have improved antitumor efficacy by delivering more drugs to tumor sites compared to free and unencapsulated therapeutics, achieving satisfactory distribution and penetration of nanoDDSs inside solid tumors, especially in stromal fibrous tumors, remains challenging. As one of the most common stromal cells in solid tumors, tumor-associated fibroblasts (TAFs) not only promote tumor growth and metastasis but also reduce the drug delivery efficiency of nanoparticles through the tumor's inherent physical and physiological barriers. Thus, TAFs have been emerging as attractive targets, and TAF-targeting nanotherapeutics have been extensively explored to enhance the tumor delivery efficiency and efficacy of various anticancer agents. The purpose of this Review is to opportunely summarize the underlying mechanisms of TAFs on obstructing nanoparticle-mediated drug delivery into tumors and discuss the current advances of a plethora of nanotherapeutic approaches for effectively targeting TAFs.
Collapse
Affiliation(s)
- Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Nicholas Little
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jonghan Park
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Cole Alexander Foster
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jiawei Chen
- Michigan Institute for Clinical & Health Research, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States.,BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States.,NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, Arizona 85721, United States.,Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
36
|
Li C, Li Z, Gong X, Liu J, Zheng T, Wang F, Wu J, Zhang B. Acidic tumor microenvironment-sensitive liposomes enhance colorectal cancer therapy by acting on both tumor cells and cancer-associated fibroblasts. NANOSCALE 2021; 13:10509-10525. [PMID: 34096957 DOI: 10.1039/d1nr01506k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in facilitating tumor invasion and metastasis, which act as the "soil" in the tumor microenvironment (TME). Accordingly, it would be a promising strategy to enhance the antitumor effect by killing both tumor cells and CAFs simultaneously. Herein, novel TME acid-responsive liposomes for co-delivery of IRI and 398 (IRI&398-s-LPs) were developed, in which the rapid release of both drugs could be triggered under acidic conditions. Notably, a CT-26/3T3 cell co-culture system was used to mimic the real TME both in vitro and in vivo. Cellular immunofluorescence revealed that IRI&398-s-LPs could efficiently decrease the activation of CAFs. In vitro cytotoxicity evaluation demonstrated that IRI&398-s-LPs exhibited higher cytotoxicity than the other liposomal formulations in the CT-26 and CT-26/3T3 cell co-culture system. In vivo NIRF imaging showed that the IRI&398-s-LPs could increase drug accumulation in the tumor sites. Furthermore, IRI&398-s-LPs not only presented superior in vivo anti-tumor activity in CT-26 bearing BALB/c mice, but also enhanced the effect in CT-26/3T3 cell bearing mice with decreased collagen and CAF biomarker expression. Furthermore, IRI&398-s-LPs also presented superior anti-metastatic efficiency in a lung metastasis model. These results indicated that this combinational strategy for eliminating both tumor cells and CAFs provides a new approach for cancer therapy, and the prepared TME-responsive liposomes for co-delivery of drugs hold promising clinical application prospects.
Collapse
Affiliation(s)
- Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Zhaohuan Li
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Xue Gong
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Jianhao Liu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Tingyue Zheng
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Fangqing Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| |
Collapse
|
37
|
Liu M, López de Juan Abad B, Cheng K. Cardiac fibrosis: Myofibroblast-mediated pathological regulation and drug delivery strategies. Adv Drug Deliv Rev 2021; 173:504-519. [PMID: 33831476 PMCID: PMC8299409 DOI: 10.1016/j.addr.2021.03.021] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/16/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis remains an unresolved problem in heart diseases. After initial injury, cardiac fibroblasts (CFs) are activated and subsequently differentiate into myofibroblasts (myoFbs) that are major mediator cells in the pathological remodeling. MyoFbs exhibit proliferative and secretive characteristics, and contribute to extracellular matrix (ECM) turnover, collagen deposition. The persistent functions of myoFbs lead to fibrotic scars and cardiac dysfunction. The anti-fibrotic treatment is hindered by the elusive mechanism of fibrosis and lack of specific targets on myoFbs. In this review, we will outline the progress of cardiac fibrosis and its contributions to the heart failure. We will also shed light on the role of myoFbs in the regulation of adverse remodeling. The communication between myoFbs and other cells that are involved in the heart injury and repair respectively will be reviewed in detail. Then, recently developed therapeutic strategies to treat fibrosis will be summarized such as i) chimeric antigen receptor T cell (CAR-T) therapy with an optimal target on myoFbs, ii) direct reprogramming from stem cells to quiescent CFs, iii) "off-target" small molecular drugs. The application of nano/micro technology will be discussed as well, which is involved in the construction of cell-based biomimic platforms and "pleiotropic" drug delivery systems.
Collapse
Affiliation(s)
- Mengrui Liu
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA.
| |
Collapse
|
38
|
Liu H, Shi Y, Qian F. Opportunities and delusions regarding drug delivery targeting pancreatic cancer-associated fibroblasts. Adv Drug Deliv Rev 2021; 172:37-51. [PMID: 33705881 DOI: 10.1016/j.addr.2021.02.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 12/17/2022]
Abstract
A dense desmoplastic stroma formed by abundant extracellular matrix and stromal cells, including cancer-associated fibroblasts (CAFs) and immune cells, is a feature of pancreatic ductal adenocarcinoma (PDAC), one of the most lethal cancer types. As the dominant cellular component of the PDAC stroma, CAFs orchestrate intensive and biologically diverse crosstalk with pancreatic cancer cells and immune cells and contribute to a unique PDAC tumor microenvironment promoting cancer proliferation, metastasis, and resistance against both chemo- and immunotherapies. Therefore, CAFs and CAF-related mechanisms have emerged as promising targets for PDAC therapy. However, several clinical setbacks and accumulating knowledge of the PDAC stroma have revealed the heterogeneity and multifaceted biological roles of CAFs, and concerns regarding "what to deliver" and "how to deliver" have arisen when designing CAF-targeted drug delivery systems to specifically inhibit tumor-supporting CAFs without impairing tumor-restricting CAFs. In this review, we will discuss the complexity of CAFs in the PDAC stroma as well as the potential opportunities and common misconceptions regarding drug delivery efforts targeting PDAC CAFs.
Collapse
Affiliation(s)
- Huiqin Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Yu Shi
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China.
| |
Collapse
|
39
|
Abstract
INTRODUCTION Compared with traditional cancer treatment methods, tumor-targeted immunotherapy can combine targeted therapy and immunotherapy with long-lasting responses to achieve synergistic therapy, which brings hope to the complete cure of cancer. AREAS COVERED This review summarizes the newest and most up-to-date advances in tumor-targeted immunotherapy, including tumor-associated macrophages (TAMs) targeted immunotherapy, regulatory T (Treg) cells targeted immunotherapy, tumor-associated fibroblasts (TAFs) targeted immunotherapy and immune checkpoints targeted immunotherapy. EXPERT OPINION Immunotherapy can restore anti-tumor immunity in the tumor microenvironment and produce a lasting immune surveillance effect. Smart multifunctional nano delivery system can effectively combine targeted therapy with immunotherapy, which has attracted extensive attention. With the deepening of research, more and more tumor-targeted immunotherapy enter into the clinical trial phases, especially antibodies and inhibitors. Tumor-targeted immunotherapy is a promising approach for conquering cancer and bringing hope for human health.
Collapse
Affiliation(s)
- Yuelin Fang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Aihua Yu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
40
|
Li H, Zhou L, Zhou J, Li Q, Ji Q. Underlying mechanisms and drug intervention strategies for the tumour microenvironment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:97. [PMID: 33722297 PMCID: PMC7962349 DOI: 10.1186/s13046-021-01893-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Cancer occurs in a complex tissue environment, and its progression depends largely on the tumour microenvironment (TME). The TME has a highly complex and comprehensive system accompanied by dynamic changes and special biological characteristics, such as hypoxia, nutrient deficiency, inflammation, immunosuppression and cytokine production. In addition, a large number of cancer-associated biomolecules and signalling pathways are involved in the above bioprocesses. This paper reviews our understanding of the TME and describes its biological and molecular characterization in different stages of cancer development. Furthermore, we discuss in detail the intervention strategies for the critical points of the TME, including chemotherapy, targeted therapy, immunotherapy, natural products from traditional Chinese medicine, combined drug therapy, etc., providing a scientific basis for cancer therapy from the perspective of key molecular targets in the TME.
Collapse
Affiliation(s)
- Haoze Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lihong Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qi Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qing Ji
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
41
|
Esmaeili J, Barati A, Ai J, Nooshabadi VT, Mirzaei Z. Employing hydrogels in tissue engineering approaches to boost conventional cancer-based research and therapies. RSC Adv 2021; 11:10646-10669. [PMID: 35423538 PMCID: PMC8695814 DOI: 10.1039/d1ra00855b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer is a complicated disease that involves the efforts of researchers to introduce and investigate novel successful treatments. Traditional cancer therapy approaches, especially chemotherapy, are prone to possible systemic side effects, such as the dysfunction of liver or kidney, neurological side effects and a decrease of bone marrow activity. Hydrogels, along with tissue engineering techniques, provide tremendous potential for scientists to overcome these issues through the release of drugs at the site of tumor. Hydrogels demonstrated competency as potent and stimulus-sensitive drug delivery systems for tumor removal, which is attributed to their unique features, including high water content, biocompatibility, and biodegradability. In addition, hydrogels have gained more attention as 3D models for easier and faster screening of cancer and tumors due to their potential in mimicking the extracellular matrix. Hydrogels as a reservoir can be loaded by an effective dosage of chemotherapeutic agents, and then deliver them to targets. In comparison to conventional procedures, hydrogels considerably decreased the total cost, duration of research, and treatment time. This study provides a general look into the potential role of hydrogels as a powerful tool to augment cancer studies for better analysis of cancerous cell functions, cell survival, angiogenesis, metastasis, and drug screening. Moreover, the upstanding application of drug delivery systems related to the hydrogel in order to sustain the release of desired drugs in the tumor cell-site were explored.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University Arak Iran
- Department of Tissue Engineering, TISSUEHUB CO. Tehran Iran
| | - Abolfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University Arak Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences Tehran 14177-55469 Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences Tehran 14177-55469 Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences Semnan Iran
| | - Zeynab Mirzaei
- Faculty of Biomedical Engineering, Amirkabir University of Technology Hafez str. 424 Tehran Iran
- Department of Tissue Engineering, TISSUEHUB CO. Tehran Iran
| |
Collapse
|
42
|
Lu G, Qiu Y, Su X. Targeting CXCL12-CXCR4 Signaling Enhances Immune Checkpoint Blockade Therapy Against Triple Negative Breast Cancer. Eur J Pharm Sci 2021; 157:105606. [PMID: 33131745 DOI: 10.1016/j.ejps.2020.105606] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/21/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Insufficient T cell infiltration in triple-negative breast cancer (TNBC) has limited its response rate to immune checkpoint blockade (ICB) therapies and motivated the development of immunostimulatory approaches to enhance the ICB therapy. CXCR4 is a chemokine receptor highly upregulated both on cell surface and cytoplasm in tumor tissues. Activating CXCR4 has been associated with increased immunosuppression in the tumor microenvironment. Here, we developed a CXCR4-targeted liposomal formulation (Liposomal-AMD3100) to enhance therapeutic efficacy of AMD3100, a CXCR4 antagonist. Particularly, AMD3100 is not only encapsulated into the liposome but coated on the surface of the formulation to serve as a targeting moiety and a dual blocker capable of inhibiting CXCR4 activation extracellularly and intracellularly. The Liposomal-AMD3100 remodeled both immune and stromal microenvironment more efficiently compared with free AMD3100, indicating better pharmacodynamic profile of AMD3100 achieved by liposomal formulation. The combination of anti-PD-L1 with Liposomal-AMD3100 formulation exhibited an increased antitumor effect and prolonged survival time compared with monotherapies in a murine TNBC model (4T1). This work proves that immune activation via liposomal delivery of CXCR4 inhibitors has a great potential to expand ICB therapies to originally ICB-insensitive cancer types.
Collapse
Affiliation(s)
- Guowen Lu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China.
| | - Yier Qiu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| | - Xiaobao Su
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| |
Collapse
|
43
|
Targeting CTGF in Cancer: An Emerging Therapeutic Opportunity. Trends Cancer 2020; 7:511-524. [PMID: 33358571 DOI: 10.1016/j.trecan.2020.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Despite the dramatic advances in cancer research over the decades, effective therapeutic strategies are still urgently needed. Increasing evidence indicates that connective tissue growth factor (CTGF), a multifunctional signaling modulator, promotes cancer initiation, progression, and metastasis by regulating cell proliferation, migration, invasion, drug resistance, and epithelial-mesenchymal transition (EMT). CTGF is also involved in the tumor microenvironment in most of the nodes, including angiogenesis, inflammation, and cancer-associated fibroblast (CAF) activation. In this review, we comprehensively discuss the expression of CTGF and its regulation, oncogenic role, clinical relevance, targeting strategies, and therapeutic agents. Herein, we propose that CTGF is a promising cancer therapeutic target that could potentially improve the clinical outcomes of cancer patients.
Collapse
|
44
|
Wu F, Xue H, Li X, Diao W, Jiang B, Wang W, Yu W, Bai J, Wang Y, Lian B, Feng W, Sun T, Qu M, Zhao C, Wang Y, Wu J, Gao Z. Enhanced targeted delivery of adenine to hepatocellular carcinoma using glycyrrhetinic acid-functionalized nanoparticles in vivo and in vitro. Biomed Pharmacother 2020; 131:110682. [PMID: 32947204 DOI: 10.1016/j.biopha.2020.110682] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a common malignancy in China and globally, is primarily treated through surgical resection and liver transplantation, with chemotherapy as a significant synergistic option. Adenine (Ade), a nucleobase, exhibits antitumor effects by blocking human hepatic carcinoma cells in S phase and inhibiting tumor cell proliferation. However, its use is limited owing to its low solubility, poor targeting ability, and nephrotoxicity. Therefore, liver-targeting drug delivery systems have attracted considerable attention for the treatment of HCC. In this study, we explored the liver-targeting efficacy and antitumor effect of adenine-loaded glycyrrhetinic acid-modified hyaluronic acid (Ade/GA-HA) nanoparticles in vitro and in vivo. The GA-HA nanoparticles possessed obvious targeting specificity toward liver cancer cells, which was mainly achieved by the specific binding of the GA ligand to the GA receptor that was highly expressed on the liver cell membrane. In vitro and in vivo results showed that Ade/GA-HA nanoparticles could inhibit liver cancer cell proliferation and migration, promote apoptosis, and significantly inhibit the growth of tumor tissues. Altogether, this study is the first to successfully demonstrate that the targeting activity and antitumor effect of Ade against HCC are enhanced by using GA-HA nanoparticles in vitro and in vivo.
Collapse
Affiliation(s)
- Fei Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Hantao Xue
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Xiaocheng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Wenbin Diao
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Bin Jiang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Weiyu Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Wenjing Yu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Yi Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Bo Lian
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Weiguo Feng
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Tongyi Sun
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Meihua Qu
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang 261041, China
| | - Chunling Zhao
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China
| | - Yubing Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China.
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China.
| | - Zhiqin Gao
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province, China.
| |
Collapse
|
45
|
Nandi T, Pradyuth S, Singh AK, Chitkara D, Mittal A. Therapeutic agents for targeting desmoplasia: current status and emerging trends. Drug Discov Today 2020; 25:S1359-6446(20)30365-2. [PMID: 32947044 DOI: 10.1016/j.drudis.2020.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/17/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Desmoplasia is a major barrier to chemotherapy in several cancers, particularly pancreatic ductal adenocarcinoma and breast cancer. Tumors comprise of cellular and noncellular components and chemoresistant cancer stem cells (CSCs) with established signaling pathways. In this review, we discuss drugs, such as pentoxifylline, aspirin, and metformin, that have been repurposed and investigated for their antidesmoplastic activity in combination with antitumor drugs. We also highlight less explored new small-molecule drugs, and gene and peptide-based therapeutics for the treatment of desmoplasia and to target CSCs. Promising results from preclinical studies have encouraged several clinical trials to evaluate these antidesmoplastic agents as adjunct to chemotherapy.
Collapse
Affiliation(s)
- Tania Nandi
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, 333031, India
| | - Sai Pradyuth
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, 333031, India
| | - Arihant Kumar Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan, 333031, India.
| |
Collapse
|
46
|
Wang J, Qian Y, Xu L, Shao Y, Zhang H, Shi F, Chen J, Cui S, Chen X, Zhu D, Hu R, Chen Z. Hyaluronic acid-shelled, peptide drug conjugate-cored nanomedicine for the treatment of hepatocellular carcinoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111261. [PMID: 32919628 DOI: 10.1016/j.msec.2020.111261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/14/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022]
Abstract
Peptide-drug conjugate (PDC) is a promising prodrug in drug delivery systems. To fabricate nanostructures with proper molecular design which can self-assemble to spherical morphologies is very important for PDC chemotherapy. In this study, a novel PDC (PDC-DOX2), in which two doxorubicin (DOX) molecules are conjugated onto a short peptide (KIGLFRWR) with self-assembly function, was designed and synthesized. PDC-DOX2 with self-assembly properties forms a spherical structure under hydrophobic interaction in water. Hyaluronic acid (HA) was then coated on PDC-DOX2 micelles to form a HA-shelled, peptide-doxorubicin conjugate-cored nanomedicine (HA@PDC-DOX2). The amount of HA can regulate the particle size and stabilization of HA@PDC-DOX2. In addition, HA can actively enhance the targeting effects of PDC-DOX2 micelles since it can interact with overexpressed receptors in cancer cells. The core-shell structured HA@PDC-DOX2 nanomedicine showed significantly enhanced potency against hepatocellular carcinoma compared to PDC-DOX2 micelles as well as free DOX. In this work, a novel PDC which can self-assemble to spherical morphologies and a core-shell structure HA@PDC-DOX2 nanomedicine are designed and prepared. It provides a convenient strategy for the size control of PDC assemblies and constructs effective PDC-based drug delivery systems for cancer treatment.
Collapse
Affiliation(s)
- Jingjing Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ying Qian
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Liu Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Yurou Shao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Hu Zhang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Fanli Shi
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jiaxin Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Siqi Cui
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Xiaoyan Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Dongwei Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Rongfeng Hu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
47
|
Single-Walled Carbon Nanohorns as Promising Nanotube-Derived Delivery Systems to Treat Cancer. Pharmaceutics 2020; 12:pharmaceutics12090850. [PMID: 32906852 PMCID: PMC7558911 DOI: 10.3390/pharmaceutics12090850] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer has become one of the most prevalent diseases worldwide, with increasing incidence in recent years. Current pharmacological strategies are not tissue-specific therapies, which hampers their efficacy and results in toxicity in healthy organs. Carbon-based nanomaterials have emerged as promising nanoplatforms for the development of targeted delivery systems to treat diseased cells. Single-walled carbon nanohorns (SWCNH) are graphene-based horn-shaped nanostructure aggregates with a multitude of versatile features to be considered as suitable nanosystems for targeted drug delivery. They can be easily synthetized and functionalized to acquire the desired physicochemical characteristics, and no toxicological effects have been reported in vivo followed by their administration. This review focuses on the use of SWCNH as drug delivery systems for cancer therapy. Their main applications include their capacity to act as anticancer agents, their use as drug delivery systems for chemotherapeutics, photothermal and photodynamic therapy, gene therapy, and immunosensing. The structure, synthesis, and covalent and non-covalent functionalization of these nanoparticles is also discussed. Although SWCNH are in early preclinical research yet, these nanotube-derived nanostructures demonstrate an interesting versatility pointing them out as promising forthcoming drug delivery systems to target and treat cancer cells.
Collapse
|
48
|
Jiang M, He K, Qiu T, Sun J, Liu Q, Zhang X, Zheng H. Tumor-targeted delivery of silibinin and IPI-549 synergistically inhibit breast cancer by remodeling the microenvironment. Int J Pharm 2020; 581:119239. [PMID: 32194211 DOI: 10.1016/j.ijpharm.2020.119239] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/06/2020] [Accepted: 03/15/2020] [Indexed: 12/12/2022]
Abstract
We induced changes in the tumor microenvironment (TME) through the synergistic actions of two drugs used in breast cancer therapy. The anti-fibrotic drug silibinin (SLB) targets tumor-associated fibroblasts and exerts immune-mediated anti-cancer effects. IPI-549, an efficient and highly selective phosphoinositide-3-kinase-gamma (PI3Kγ) inhibitor, was applied to alter the balance of immunosuppressive cells by inhibiting PI3Kγ molecules; it also promotes anti-tumor immunity. We developed nanoparticle formulations to encapsulate both drugs into the targeting carrier aminoethyl anisamide-polyethylene glycol-polycaprolactone (AEAA-PEG-PCL) respectively. The drugs were intravenously delivered in mice and resulted in an increase in anti-tumor efficacy and apoptotic tumor tissue compared with either IPI-549 or SLB alone in 4T1 breast cancer cell-derived tumors. Furthermore, a significant reduction in regulatory T (Treg) cells and myeloid suppressor cells (MDSCs) was observed. A normalized TME structure was also observed, including angiogenesis suppression, antifibrotic effects and the inhibition of collagen formation in the tumor tissue, significantly enhancing the anti-tumor effects. In summary, this combination strategy may offer an alternative treatment for breast cancer.
Collapse
Affiliation(s)
- Min Jiang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Kaiyong He
- Hubei Institute for Drug Control, Wuhan 430070, China
| | - Tong Qiu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Jiahui Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Xueqiong Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| | - Hua Zheng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
49
|
Guo J, Zeng H, Chen Y. Emerging Nano Drug Delivery Systems Targeting Cancer-Associated Fibroblasts for Improved Antitumor Effect and Tumor Drug Penetration. Mol Pharm 2020; 17:1028-1048. [PMID: 32150417 DOI: 10.1021/acs.molpharmaceut.0c00014] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Huating Zeng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
50
|
Mortezaee K. CXCL12/CXCR4 axis in the microenvironment of solid tumors: A critical mediator of metastasis. Life Sci 2020; 249:117534. [PMID: 32156548 DOI: 10.1016/j.lfs.2020.117534] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/24/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Tumors are dynamic tissue masses, so requiring continuous exposure to the host cells, nurturing them into pave a path for tumor growth and metastasis. C-X-C chemokine ligand 12 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) is the key signaling for such aim. Gathering knowledge about the activity within this axis would deepen our insight into the utmost importance this signaling taken to attract and cross-connect multiple cells within the tumor microenvironment (TME) aiming for tumor progression and metastasis. The concept behind this review is to underscore the multi-tasking roles taken by CXCL12/CXCR4 signaling in tumor metastasis, and to also suggest some strategies to target the activities within this axis.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|