1
|
Yu F, Zhang G, Sun J, Zhao Y, Qi Y, Han X, Ai C, Sun W, Duan J, Yu D. Nanotension Relief Agent Enhances Tissue Penetration by Reducing Solid Stress in Pancreatic Ductal Adenocarcinoma via Rho/ROCK Pathway Inhibition. Biomater Res 2025; 29:0173. [PMID: 40207257 PMCID: PMC11979343 DOI: 10.34133/bmr.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
The formidable contractile tension exerted by cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinoma (PDAC) tissue is crucial for maintaining high tissue solid stress (TSS), which impedes the delivery and penetration of chemotherapeutic drugs. To address this obstacle, we constructed a pH-responsive nanotension relief agent (FS@MMS), in which fasudil (FS) was ingeniously conjugated to mesoporous silica encapsulated with magnetic iron oxide (MMS). The nanotension relief agent was demonstrated to inhibit the synthesis of phosphorylated myosin light chain by blocking the Rho/Rho-associated serine/threonine kinase (ROCK) pathway, triggering the swift transformation of high-tension CAFs into low-tension CAFs in PDAC tissue, which relieves TSS and enhances drug penetration in Panc02/NIH-3T3 multicellular tumor spheroids. When the nanotension relief agent was further loaded with the chemotherapeutic drug gemcitabine (GEM), as FS@MMS-GEM, the enhanced permeation of GEM progressively killed tumor cells and amplified their TSS-relief properties, thereby maximizing the anticancer efficacy of chemotherapeutic agents in Panc02/NIH-3T3 coplanted model mice. The magnetic resonance imaging results revealed that the synergistic effect substantially improved drug delivery and penetration efficiency. The developed approach holds great potential for improving chemotherapy efficacy in PDAC and provides a novel therapeutic approach for the treatment of related stroma-rich tumors.
Collapse
Affiliation(s)
- Feiran Yu
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Gaorui Zhang
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jintang Sun
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yuxuan Zhao
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yafei Qi
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiaoyu Han
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chen Ai
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Weikai Sun
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiazhi Duan
- Institute for Advanced Interdisciplinary Research,
University of Jinan, Jinan 250022, China
| | - Dexin Yu
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
| |
Collapse
|
2
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Muheyati M, Wu G, Li Y, Pan Z, Chen Y. Supramolecular nanotherapeutics based on cucurbiturils. J Nanobiotechnology 2024; 22:790. [PMID: 39710716 DOI: 10.1186/s12951-024-03024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024] Open
Abstract
Polymeric biomaterials have important applications in aiding clinical disease treatment, including drug delivery, bioimaging, and tissue engineering. Currently, conventional tumor chemotherapy faces obstacles such as poor solubility/stability, inability to target, and uncontrolled drug release in clinical trials, for which the emergence of supramolecular material therapeutics combining non-covalent interactions with conventional therapies is a very promising candidate. Due to their molecular recognition abilities with a range of biomolecules, cucurbit[n]uril (CB[n]), a type of macrocyclic receptors with robust backbones, hydrophobic cavities, and carbonyl-binding channels, have garnered a lot of attention. Therefore, this paper reviews recent advances in CB[n] material-based supramolecular therapeutics for clinical treatments, including targeted delivery applications and related imaging and sensing systems. This study also covers the distinctive benefits of CB materials for biological applications, as well as the trends and prospects of this interdisciplinary subject, based on numerous state-of-the-art research findings.
Collapse
Affiliation(s)
- Maiyier Muheyati
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Guangheng Wu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yilin Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Ziting Pan
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, People's Republic of China
- School of Basic Medicine, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
4
|
Carlos A, Mendes M, Cruz MT, Pais A, Vitorino C. Ferroptosis driven by nanoparticles for tackling glioblastoma. Cancer Lett 2024; 611:217392. [PMID: 39681210 DOI: 10.1016/j.canlet.2024.217392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Glioblastoma (GBM) is the most aggressive, malignant, and drug-resistant brain tumor. There are no effective treatment options for GBM, which usually leads to relapses that cause patients to die a few months later. Ferroptosis, a newly discovered mechanism of regulated cell death, has been identified as a tumor suppressor in solid tumors and represents an alternative to apoptosis resistance. This mechanism of cell death is characterized by iron overload, which is responsible for generating reactive oxygen species (ROS) in the cell. Understanding the ferroptosis pathway and its key regulators can be used to develop rational delivery systems that specifically target these regulators in GBM cells and promote cell death. This review conducted a systematic literature search to better understand the potential of ferroptosis as a target for developing nanoparticles to tackle GBM. The mechanisms of action, design parameters, efficacy, and safety concerns of 16 nanoparticles were evaluated, demonstrating the potential of combining ferroptosis inducers with nanocarriers to promote a selective delivery to the tumor microenvironment.
Collapse
Affiliation(s)
- Ana Carlos
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Departmente of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Neurosciences and Cell Biology (CNC) and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Departmente of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Departmente of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
5
|
Abbasi S, Khademi S, Montazerabadi A, Sahebkar A. FAP-Targeted Nanoparticle-based Imaging in Cancer: A Systematic Review. J Biomed Phys Eng 2024; 14:323-334. [PMID: 39175559 PMCID: PMC11336055 DOI: 10.31661/jbpe.v0i0.2404-1754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/12/2024] [Indexed: 08/24/2024]
Abstract
Background Fibroblast Activation Protein (FAP)-targeted nanoparticles (NPs) are designed to accumulate in cancerous stroma. These NPs hold promise for imaging applications in cancer therapy. Objective This systematic review aimed to comprehensively explore the use of FAP-targeting NPs for cancer diagnosis through different imaging modalities. Material and Methods This systematic review followed the framework proposed by O'Malley and Arksey. Peer-reviewed studies were searched in the Scopus, Science Direct, PubMed, and Google Scholar databases. Eligible studies were selected, and data were extracted to investigate the FAP-targeting NPs in imaging. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline was also utilized to present the results. Results Five studies met the specified inclusion criteria and were finally selected for analysis. The extracted data was classified into two categories: general and specific data. The general group indicated that most studies have been conducted in Mexico and have increased since 2022, and the specific group showed that colorectal cancer and Nude mice have received the most research attention. Furthermore, FAP-targeted NPs have demonstrated superior diagnostic imaging capabilities, even compared to specific methods for each cancer type. Also, they have been safe, with no toxicity. Conclusion FAP-targeted NPs using different ligands, such as Fibroblast Activation Protein Inhibitor (FAPI), can accurately detect tumors and metastases, and outperform specific cancer peptides like PSMA in cancer diagnosis. They are also non-toxic and do not cause radiation damage to tissues. Therefore, FAP-targeted NPs have the potential to serve as a viable alternative to FAP-targeted radionuclides for cancer diagnosis.
Collapse
Affiliation(s)
- Samaneh Abbasi
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Khademi
- Department of Radiology Technology, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Montazerabadi
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Jia N, Wang Q, Li W, Chen D, Hu H. Membrane Fusion Liposomes Deliver Antifibrotic and Chemotherapeutic Drugs Sequentially to Enhance Tumor Treatment Efficacy by Reshaping Tumor Microenvironment. Adv Healthc Mater 2024; 13:e2400219. [PMID: 38657266 DOI: 10.1002/adhm.202400219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/14/2024] [Indexed: 04/26/2024]
Abstract
The intricate tumor microenvironment in triple-negative breast cancer (TNBC) hampers chemotherapy and immunotherapy efficacy due to dense extracellular matrix (ECM) by tumor-associated fibroblasts (TAFs). Nanoparticle-based therapies, especially "all-in-one" nanoparticles, have shown great potential in combined drug delivery strategies to reshape the tumor microenvironment and enhance therapeutic efficiency. However, these "all-in-one" nanoparticles suffer from limitations in targeting different target cells, uncontrollable dosing ratio, and disregarding the impact of delivery schedules. This study prepared cell membrane fusion liposomes (TAFsomes and CCMsomes) to load FDA-approved antifibrotic drug pirfenidone (PFD/TAFsomes) and antitumor drug doxorubicin (DOX/CCMsomes). These liposomes can specifically target TAFs cells and tumor cells, and combined administration can effectively inhibit TAFs activity, reshape the tumor microenvironment (TME), and significantly enhance the tumor chemotherapy efficacy. Combined drug delivery defeats "all-in-one" liposomes (DOX/PFD/Liposomes, DOX/PFD/TAFsomes, and DOX/PFD/CCMsomes) by flexibly adjusting the drug delivery ratio. Moreover, an asynchronous delivery strategy that optimizes the administration schedule not only further improves the therapeutic effect, but also amplifies the effectiveness of α-PD-L1 immunotherapy by modulating the tumor immune microenvironment. This delivery strategy provides a personalized treatment approach with clinical translation potential, providing new ideas for enhancing the therapeutic effect against solid tumors such as TNBC.
Collapse
Affiliation(s)
- Nan Jia
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Qi Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Wenpan Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| |
Collapse
|
7
|
Li J, He H, Liu S, Li X, Wu F. Revealing tumor cells and tissues with high selectivity through folic acid-targeted nanofluorescence probes responsive to acidic microenvironments. Front Oncol 2024; 14:1404148. [PMID: 38933449 PMCID: PMC11199542 DOI: 10.3389/fonc.2024.1404148] [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: 03/20/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor-specific fluorescent probes must fulfill the dual requirements of targeted accumulation within tumors and high-resolution imaging capabilities. To achieve both tumor-targeted accumulation and high-resolution imaging performance, we developed a composite comprising an acid-responsive bodipy conjugated to amphiphilic PEG-b-PLA polymer, along with folic acid (FA)-modified PEG-b-PLA as a targeting moiety for active tumor-specific accumulation. Finally, a novel assembly of hybrid fluorescent nanoparticles was successfully synthesized by integrating these two components, demonstrating exceptional responsiveness to acidic conditions for fluorescence excitation and remarkable tumor-targeted accumulation capabilities. We conducted comprehensive in vitro and in vivo investigations employing techniques such as analysis of physicochemical properties, fluorescence-based probes detection at varying pH levels, assessment of in vitro cytotoxicity, evaluation of cellular uptake capacity, analysis of lysosomal co-localization imaging, examination of tumor fluorescence images in vivo, and investigation of biological distribution patterns. The results demonstrated that the acid-responsive nanofluorescence probe we designed and synthesized possesses desirable physical and chemical characteristics, including a small particle size and low cytotoxicity. Moreover, it exhibits rapid real-time response to acidic environments and displays enhanced fluorescence intensity, enabling the real-time tracking of probe entry into tumor cells as well as intracellular lysozyme accumulation. We achieved highly specific in vivo tumor visualization by combining nanoprobes targeting folate receptor. Through imaging cervical tumor mice, we demonstrated the precise imaging performance and high targeted accumulation of FA-targeted nanofluorescence probes in tumor tissue. Furthermore, we confirmed the in vivo safety of the FA-targeted nanofluorescence probe through biological distribution analysis. These findings highlight the potential widespread application of FA-targeted acid-responsive nanofluorescence probes for selective imaging of tumor cells and tissues.
Collapse
Affiliation(s)
- Jing Li
- Neurobiology Laboratory, Wannan Medical College, Wuhu, China
| | - Hongyi He
- College of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Shuyan Liu
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, China
| | - Xining Li
- School of Medicine, Huzhou University, Huzhou, China
| | - Fengfeng Wu
- Department of Orthopedics and Rehabilitation, Huzhou Hospital of Zhejiang University School of Medicine, Huzhou, China
- Department of Orthopedics and Rehabilitation, Huzhou Central Hospital, Huzhou, China
| |
Collapse
|
8
|
De Rubis G, Chakraborty A, Paudel KR, Wang C, Kannaujiya V, Wich PR, Hansbro PM, Samuel CS, Oliver B, Dua K. Exploring the anti-inflammatory and anti-fibrotic activity of NFκB decoy oligodeoxynucleotide-loaded spermine-functionalized acetalated nanoparticles. Chem Biol Interact 2024; 396:111059. [PMID: 38761875 DOI: 10.1016/j.cbi.2024.111059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Chronic inflammation, oxidative stress, and airway remodelling represent the principal pathophysiological features of chronic respiratory disorders. Inflammation stimuli like lipopolysaccharide (LPS) activate macrophages and dendritic cells, with concomitant M1 polarization and release of pro-inflammatory cytokines. Chronic inflammation and oxidative stress lead to airway remodelling causing irreversible functional and structural alterations of the lungs. Airway remodelling is multifactorial, however, the hormone transforming growth factor-β (TGF-β) is one of the main contributors to fibrotic changes. The signalling pathways mediating inflammation and remodelling rely both on the transcription factor nuclear factor-κB (NFκB), underlying the potential of NFκB inhibition as a therapeutic strategy for chronic respiratory disorders. In this study, we encapsulated an NFκB-inhibiting decoy oligodeoxynucleotide (ODN) in spermine-functionalized acetalated dextran (SpAcDex) nanoparticles and tested the in vitro anti-inflammatory and anti-remodelling activity of this formulation. We show that NF-κB ODN nanoparticles counteract inflammation by reversing LPS-induced expression of the activation marker CD40 in myeloid cells and counteracts remodelling features by reversing the TGF-β-induced expression of collagen I and α-smooth muscle actin in human dermal fibroblast. In summary, our study highlights the great potential of inhibiting NFκB via decoy ODN as a therapeutic strategy tackling multiple pathophysiological features underlying chronic respiratory conditions.
Collapse
Affiliation(s)
- Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Amlan Chakraborty
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK; Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia.
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, 2007, Australia
| | - Chao Wang
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Vinod Kannaujiya
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Peter Richard Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, 2007, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Brian Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
9
|
Yadav P, Rana K, Nardini V, Khan A, Pani T, Kar A, Jain D, Chakraborty R, Singh R, Jha SK, Mehta D, Sharma H, Sharma RD, Deo SVS, Sengupta S, Patil VS, Faccioli LH, Dasgupta U, Bajaj A. Engineered nanomicelles inhibit the tumour progression via abrogating the prostaglandin-mediated immunosuppression. J Control Release 2024; 368:548-565. [PMID: 38462044 DOI: 10.1016/j.jconrel.2024.03.009] [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: 07/30/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Cancer treatment is challenged due to immunosuppressive inflammatory tumour microenvironment (TME) caused by infiltration of tumour-promoting and inhibition of tumour-inhibiting immune cells. Here, we report the engineering of chimeric nanomicelles (NMs) targeting the cell proliferation using docetaxel (DTX) and inflammation using dexamethasone (DEX) that alters the immunosuppressive TME. We show that a combination of phospholipid-DTX conjugate and PEGylated-lipid-DEX conjugate can self-assemble to form sub-100 nm chimeric NMs (DTX-DEX NMs). Anti-cancer activities against syngeneic and xenograft mouse models showed that the DTX-DEX NMs are more effective in tumour regression, enhance the survival of mice over other treatment modes, and alter the tumour stroma. DTX-DEX NMs cause a significant reduction in myeloid-derived suppressor cells, alter the polarization of macrophages, and enhance the accumulation of cytotoxic CD4+ and CD8+ T cells in tumour tissues, along with alterations in cytokine expression. We further demonstrated that these DTX-DEX NMs inhibit the synthesis of prostaglandins, especially PGE2, by targeting the cyclooxygenase 2 that is partly responsible for immunosuppressive TME. Therefore, this study presents, for the first time, the engineering of lithocholic acid-derived chimeric NMs that affect the prostaglandin pathway, alter the TME, and mitigate tumour progression with enhanced mice survival.
Collapse
Affiliation(s)
- Poonam Yadav
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Kajal Rana
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Viviani Nardini
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av do Café, s.n, Ribeirão Preto 14040-903, SP, Brazil
| | - Ali Khan
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Trishna Pani
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Animesh Kar
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Dolly Jain
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Ruchira Chakraborty
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Ragini Singh
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Somesh K Jha
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Devashish Mehta
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Harsh Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Ravi Datta Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - S V S Deo
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sagar Sengupta
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India; National Institute of Biomedical Genomics, Post office- Netaji Subhas Sanatorium, Kalyani 741251, India
| | - Veena S Patil
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Lúcia Helena Faccioli
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av do Café, s.n, Ribeirão Preto 14040-903, SP, Brazil
| | - Ujjaini Dasgupta
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India.
| |
Collapse
|
10
|
Xiang Y, Chen Q, Nan Y, Liu M, Xiao Z, Yang Y, Zhang J, Ying X, Long X, Wang S, Sun J, Huang Q, Ai K. Nitric Oxide‐Based Nanomedicines for Conquering TME Fortress: Say “NO” to Insufficient Tumor Treatment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202312092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Indexed: 01/02/2025]
Abstract
AbstractAlmost all cancer treatments are significantly limited by the strong tumor microenvironment (TME) fortress formed by abnormal vasculature, dense extracellular matrix (ECM), multidrug resistance (MDR) system, and immune “cold” environment. In the huge efforts of dismantling the TME fortress, nitric oxide (NO)‐based nanomedicines are increasingly occupying a central position and have already been identified as super “strong polygonal warriors” to dismantle TME fortress for efficient cancer treatment, benefiting from NO's unique physicochemical properties and extremely fascinating biological effects. However, there is a paucity of systematic review to elaborate on the progress and fundamental mechanism of NO‐based nanomedicines in oncology from this aspect. Herein, the key characteristics of TME fortress and the potential of NO in reprogramming TME are delineated and highlighted. The evolution of NO donors and the advantages of NO‐based nanomedicines are discussed subsequently. Moreover, the latest progress of NO‐based nanomedicines for solid tumors is comprehensively reviewed, including normalizing tumor vasculature, overcoming ECM barrier, reversing MDR, and reactivating the immunosuppression TME. Lastly, the prospects, limitations, and future directions on NO‐based nanomedicines for TME manipulation are discussed to provide new insights into the construction of more applicable anticancer nanomedicines.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yayun Nan
- Geriatric Medical Center People's Hospital of Ningxia Hui Autonomous Region Yinchuan Ningxia 750002 P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yuqi Yang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Jinping Zhang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Xingyu Long
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Jian Sun
- College of Pharmacy Xinjiang Medical University Urumqi 830017 P. R. China
| | - Qiong Huang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and Treatment Ministry of Education Xiangya Hospital Central South University Changsha 410078 P. R. China
| |
Collapse
|
11
|
Ghosh A, Maske P, Patel V, Dubey J, Aniket K, Srivastava R. Theranostic applications of peptide-based nanoformulations for growth factor defective cancers. Int J Biol Macromol 2024; 260:129151. [PMID: 38181914 DOI: 10.1016/j.ijbiomac.2023.129151] [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: 07/01/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
Growth factors play a pivotal role in orchestrating cellular growth and division by binding to specific cell surface receptors. Dysregulation of growth factor production or activity can contribute to the uncontrolled cell proliferation observed in cancer. Peptide-based nanoformulations (PNFs) have emerged as promising therapeutic strategies for growth factor-deficient cancers. PNFs offer multifaceted capabilities including targeted delivery, imaging modalities, combination therapies, resistance modulation, and personalized medicine approaches. Nevertheless, several challenges remain, including limited specificity, stability, pharmacokinetics, tissue penetration, toxicity, and immunogenicity. To address these challenges and optimize PNFs for clinical translation, in-depth investigations are warranted. Future research should focus on elucidating the intricate interplay between peptides and nanoparticles, developing robust spectroscopic and computational methodologies, and establishing a comprehensive understanding of the structure-activity relationship governing peptide-nanoparticle interactions. Bridging these knowledge gaps will propel the translation of peptide-nanoparticle therapies from bench to bedside. While a few peptide-nanoparticle drugs have obtained FDA approval for cancer treatment, the integration of nanostructured platforms with peptide-based medications holds tremendous potential to expedite the implementation of innovative anticancer interventions. Therefore, growth factor-deficient cancers present both challenges and opportunities for targeted therapeutic interventions, with peptide-based nanoformulations positioned as a promising avenue. Nonetheless, concerted research and development endeavors are essential to optimize the specificity, stability, and safety profiles of PNFs, thereby advancing the field of peptide-based nanotherapeutics in the realm of oncology research.
Collapse
Affiliation(s)
- Arnab Ghosh
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India.
| | - Priyanka Maske
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India
| | - Vinay Patel
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India
| | - Jyoti Dubey
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India
| | - Kundu Aniket
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India.
| | - Rohit Srivastava
- Indian Institute of Technology Bombay, NanoBios lab, Department of Biosciences and Bioengineering, Mumbai, India.
| |
Collapse
|
12
|
Chen J, Chen R, Chau CV, Sedgwick AC, Xue Q, Chen T, Zeng S, Chen N, Wong KKY, Song L, Ren Y, Yang J, Sessler JL, Liu C. Targeted Cyclo[8]pyrrole-Based NIR-II Photoacoustic Tomography Probe for Suppression of Orthotopic Pancreatic Tumor Growth and Intra-abdominal Metastases. J Am Chem Soc 2024; 146:4620-4631. [PMID: 38330912 DOI: 10.1021/jacs.3c11666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Pancreatic cancer is highly lethal. New diagnostic and treatment modalities are desperately needed. We report here that an expanded porphyrin, cyclo[8]pyrrole (CP), with a high extinction coefficient (89.16 L/g·cm) within the second near-infrared window (NIR-II), may be formulated with an αvβ3-specific targeting peptide, cyclic-Arg-Gly-Asp (cRGD), to form cRGD-CP nanoparticles (cRGD-CPNPs) with promising NIR-II photothermal (PT) therapeutic and photoacoustic (PA) imaging properties. Studies with a ring-array PA tomography system, coupled with analysis of control nanoparticles lacking a targeting element (CPNPs), revealed that cRGD conjugation promoted the delivery of the NPs through abnormal vessels around the tumor to the solid tumor core. This proved true in both subcutaneous and orthotopic pancreatic tumor mice models, as confirmed by immunofluorescent studies. In combination with NIR-II laser photoirradiation, the cRGD-CPNPs provided near-baseline tumor growth inhibition through PTT both in vitro and in vivo. Notably, the combination of the present cRGD-CPNPs and photoirradiation was found to inhibit intra-abdominal metastases in an orthotopic pancreatic tumor mouse model. The cRGD-CPNPs also displayed good biosafety profiles, as inferred from PA tomography, blood analyses, and H&E staining. They thus appear promising for use in combined PA imaging and PT therapeutic treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jingqin Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rui Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Calvin V Chau
- Department of Chemistry, University of Texas at Austin, 105 East 24th Street A5300, Austin, Texas 78712-1224, United States
| | - Adam C Sedgwick
- Department of Chemistry, Kings College London, 7 Trinity Street, London SE1 1DB, U.K
| | - Qiang Xue
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tao Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Silue Zeng
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ningbo Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 999077, China
| | - Kenneth K Y Wong
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 999077, China
| | - Liang Song
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yaguang Ren
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jian Yang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China
| | - Jonathan L Sessler
- Department of Chemistry, University of Texas at Austin, 105 East 24th Street A5300, Austin, Texas 78712-1224, United States
| | - Chengbo Liu
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
13
|
Cecchi D, Jackson N, Beckham W, Chithrani DB. Improving the Efficacy of Common Cancer Treatments via Targeted Therapeutics towards the Tumour and Its Microenvironment. Pharmaceutics 2024; 16:175. [PMID: 38399237 PMCID: PMC10891984 DOI: 10.3390/pharmaceutics16020175] [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: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer is defined as the uncontrolled proliferation of heterogeneous cell cultures in the body that develop abnormalities and mutations, leading to their resistance to many forms of treatment. Left untreated, these abnormal cell growths can lead to detrimental and even fatal complications for patients. Radiation therapy is involved in around 50% of cancer treatment workflows; however, it presents significant recurrence rates and normal tissue toxicity, given the inevitable deposition of the dose to the surrounding healthy tissue. Chemotherapy is another treatment modality with excessive normal tissue toxicity that significantly affects patients' quality of life. To improve the therapeutic efficacy of radiotherapy and chemotherapy, multiple conjunctive modalities have been proposed, which include the targeting of components of the tumour microenvironment inhibiting tumour spread and anti-therapeutic pathways, increasing the oxygen content within the tumour to revert the hypoxic nature of the malignancy, improving the local dose deposition with metal nanoparticles, and the restriction of the cell cycle within radiosensitive phases. The tumour microenvironment is largely responsible for inhibiting nanoparticle capture within the tumour itself and improving resistance to various forms of cancer therapy. In this review, we discuss the current literature surrounding the administration of molecular and nanoparticle therapeutics, their pharmacokinetics, and contrasting mechanisms of action. The review aims to demonstrate the advancements in the field of conjugated nanomaterials and radiotherapeutics targeting, inhibiting, or bypassing the tumour microenvironment to promote further research that can improve treatment outcomes and toxicity rates.
Collapse
Affiliation(s)
- Daniel Cecchi
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
| | - Nolan Jackson
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
| | - Wayne Beckham
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
- British Columbia Cancer-Victoria, Victoria, BC V8R 6V5, Canada
| | - Devika B. Chithrani
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (D.C.)
- Centre for Advanced Materials and Related Technologies, Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- Department of Computer Science, Mathematics, Physics and Statistics, Okanagan Campus, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
14
|
Yuan D, Zheng BW, Zheng BY, Niu HQ, Zou MX, Liu SL, Liu FS. Global cluster analysis and network visualization in cancer-associated fibroblast: insights from Web of Science database from 1999 to 2021. Eur J Med Res 2023; 28:549. [PMID: 38031121 PMCID: PMC10685623 DOI: 10.1186/s40001-023-01527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND A scientific and comprehensive analysis of the current status and trends in the field of cancer-associated fibroblast (CAF) research is worth investigating. This study aims to investigate and visualize the development, research frontiers, and future trends in CAFs both quantitatively and qualitatively based on a bibliometric approach. METHODS A total of 5518 publications were downloaded from the Science Citation Index Expanded of Web of Science Core Collection from 1999 to 2021 and identified for bibliometric analysis. Visualized approaches, OriginPro (version 9.8.0.200) and R (version 4.2.0) software tools were used to perform bibliometric and knowledge-map analysis. RESULTS The number of publications on CAFs increased each year, and the same tendency was observed in the RRI. Apart from China, the countries with the largest number of publications and the most cited frequency were mainly Western developed countries, especially the USA. Cancers was the journal with the largest number of articles published in CAFs, and Oncology was the most popular research orientation. The most productive author was Lisanti MP, and the University of Texas System was ranked first in the institutions. In addition, the topics of CAFs could be divided into five categories, including tumor classification, prognostic study, oncologic therapies, tumor metabolism and tumor microenvironment. CONCLUSIONS This is the first thoroughly scientific bibliometric analysis and visualized study of the global research field on CAFs over the past 20 years. The study may provide benefits for researchers to master CAFs' dynamic evolution and research trends.
Collapse
Affiliation(s)
- Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bo-Wen Zheng
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Bo-Yv Zheng
- Department of Orthopedics Surgery, General Hospital of the Central Theater Command, Wuhan, 430061, China
| | - Hua-Qing Niu
- Department of Ophthalmology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, China
| | - Ming-Xiang Zou
- Department of Spine Surgery, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
| | - Song-Lin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Fu-Sheng Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
15
|
Bhattacharya T, Preetam S, Ghosh B, Chakrabarti T, Chakrabarti P, Samal SK, Thorat N. Advancement in Biopolymer Assisted Cancer Theranostics. ACS APPLIED BIO MATERIALS 2023; 6:3959-3983. [PMID: 37699558 PMCID: PMC10583232 DOI: 10.1021/acsabm.3c00458] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
Applications of nanotechnology have increased the importance of research and nanocarriers, which have revolutionized the method of drug delivery to treat several diseases, including cancer, in the past few years. Cancer, one of the world's fatal diseases, has drawn scientists' attention for its multidrug resistance to various chemotherapeutic drugs. To minimize the side effects of chemotherapeutic agents on healthy cells and to develop technological advancement in drug delivery systems, scientists have developed an alternative approach to delivering chemotherapeutic drugs at the targeted site by integrating it inside the nanocarriers like synthetic polymers, nanotubes, micelles, dendrimers, magnetic nanoparticles, quantum dots (QDs), lipid nanoparticles, nano-biopolymeric substances, etc., which has shown promising results in both preclinical and clinical trials of cancer management. Besides that, nanocarriers, especially biopolymeric nanoparticles, have received much attention from researchers due to their cost-effectiveness, biodegradability, treatment efficacy, and ability to target drug delivery by crossing the blood-brain barrier. This review emphasizes the fabrication processes, the therapeutic and theragnostic applications, and the importance of different biopolymeric nanocarriers in targeting cancer both in vitro and in vivo, which conclude with the challenges and opportunities of future exploration using biopolymeric nanocarriers in onco-therapy with improved availability and reduced toxicity.
Collapse
Affiliation(s)
- Tanima Bhattacharya
- Department
of Food and Nutrition, College of Human Ecology, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic
of Korea
- Nondestructive
Bio-Sensing Laboratory, Dept. of Biosystems Machinery Engineering,
College of Agriculture and Life Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Subham Preetam
- Centre
for Biotechnology, Siksha O Anusandhan (Deemed
to be University), Bhubaneswar 751024, Odisha, India
- Daegu
Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | - Basab Ghosh
- KIIT
School of Biotechnology, Kalinga Institute
of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Tulika Chakrabarti
- Department
of Chemistry, Sir Padampat Singhania University, Bhatewar, Udaipur 313601, Rajasthan, India
| | | | - Shailesh Kumar Samal
- Section of
Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Nanasaheb Thorat
- Nuffield
Department of Women’s & Reproductive Health, Medical Science
Division, John Radcliffe Hospital University
of Oxford, Oxford OX3 9DU, United Kingdom
- Department
of Physics, Bernal Institute and Limerick Digital Cancer Research
Centre (LDCRC), University of Limerick, Castletroy, Limerick V94T9PX, Ireland
| |
Collapse
|
16
|
Gubert P, Gubert G, de Oliveira RC, Fernandes ICO, Bezerra IC, de Ramos B, de Lima MF, Rodrigues DT, da Cruz AFN, Pereira EC, Ávila DS, Mosca DH. Caenorhabditis elegans as a Prediction Platform for Nanotechnology-Based Strategies: Insights on Analytical Challenges. TOXICS 2023; 11:239. [PMID: 36977004 PMCID: PMC10059662 DOI: 10.3390/toxics11030239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Nanotechnology-based strategies have played a pivotal role in innovative products in different technological fields, including medicine, agriculture, and engineering. The redesign of the nanometric scale has improved drug targeting and delivery, diagnosis, water treatment, and analytical methods. Although efficiency brings benefits, toxicity in organisms and the environment is a concern, particularly in light of global climate change and plastic disposal in the environment. Therefore, to measure such effects, alternative models enable the assessment of impacts on both functional properties and toxicity. Caenorhabditis elegans is a nematode model that poses valuable advantages such as transparency, sensibility in responding to exogenous compounds, fast response to perturbations besides the possibility to replicate human disease through transgenics. Herein, we discuss the applications of C. elegans to nanomaterial safety and efficacy evaluations from one health perspective. We also highlight the directions for developing appropriate techniques to safely adopt magnetic and organic nanoparticles, and carbon nanosystems. A description was given of the specifics of targeting and treatment, especially for health purposes. Finally, we discuss C. elegans potential for studying the impacts caused by nanopesticides and nanoplastics as emerging contaminants, pointing out gaps in environmental studies related to toxicity, analytical methods, and future directions.
Collapse
Affiliation(s)
- Priscila Gubert
- Keizo Asami Institute, iLIKA, Federal University of Pernambuco, Recife 50670-901, Brazil
- Graduate Program in Biology Applied to Health, PPGBAS, Federal University of Pernambuco, Recife 50670-901, Brazil
- Graduate Program in Pure and Applied Chemistry, POSQUIPA, Federal University of Western of Bahia, Bahia 47808-021, Brazil
| | - Greici Gubert
- Postdoctoral Program in Chemistry, Federal University of São Carlos, São Carlos 13565-905, Brazil
| | | | - Isabel Cristina Oliveira Fernandes
- Keizo Asami Institute, iLIKA, Federal University of Pernambuco, Recife 50670-901, Brazil
- Graduate Program in Biology Applied to Health, PPGBAS, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | - Bruna de Ramos
- Oceanography Department, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Milena Ferreira de Lima
- Keizo Asami Institute, iLIKA, Federal University of Pernambuco, Recife 50670-901, Brazil
- Graduate Program in Biology Applied to Health, PPGBAS, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Daniela Teixeira Rodrigues
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria 97105-900, Brazil
| | | | - Ernesto Chaves Pereira
- Postdoctoral Program in Chemistry, Federal University of São Carlos, São Carlos 13565-905, Brazil
| | - Daiana Silva Ávila
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria 97105-900, Brazil
- Graduate Program in Biochemistry, Federal University of Pampa (UNIPAMPA), Uruguaiana 97501-970, Brazil
| | - Dante Homero Mosca
- Postdoctoral Program in Physics, Federal University of Paraná, Curitiba 80060-000, Brazil
| |
Collapse
|
17
|
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
|
18
|
Bae I, Kim TG, Kim T, Kim D, Kim DH, Jo J, Lee YJ, Jeong YI. Phenethyl Isothiocyanate-Conjugated Chitosan Oligosaccharide Nanophotosensitizers for Photodynamic Treatment of Human Cancer Cells. Int J Mol Sci 2022; 23:13802. [PMID: 36430279 PMCID: PMC9693342 DOI: 10.3390/ijms232213802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The aim of this study is to synthesize phenethyl-conjugated chitosan oligosaccharide (COS) (abbreviated as ChitoPEITC) conjugates and then fabricate chlorin E6 (Ce6)-incorporated nanophotosensitizers for photodynamic therapy (PDT) of HCT-116 colon carcinoma cells. PEITC was conjugated with the amine group of COS. Ce6-incorporated nanophotosensitizers using ChitoPEITC (ChitoPEITC nanophotosensitizers) were fabricated by dialysis method. 1H nuclear magnetic resonance (NMR) spectra showed that specific peaks of COS and PEITC were observed at ChitoPEITC conjugates. Transmission electron microscope (TEM) confirmed that ChitoPEITC nanophotosensitizers have spherical shapes with small hydrodynamic diameters less than 200 nm. The higher PEITC contents in the ChitoPEITC copolymer resulted in a slower release rate of Ce6 from nanophotosensitizers. Furthermore, the higher Ce6 contents resulted in a slower release rate of Ce6. In cell culture study, ChitoPEITC nanophotosensitizers showed low toxicity against normal CCD986Sk human skin fibroblast cells and HCT-116 human colon carcinoma cells in the absence of light irradiation. ChitoPEITC nanophotosensitizers showed a significantly higher Ce6 uptake ratio than that of free Ce6. Under light irradiation, cellular reactive oxygen species (ROS) production of nanophotosensitizers was significantly higher than that of free Ce6. Especially, PEITC and/or ChitoPEITC themselves contributed to the production of cellular ROS regardless of light irradiation. ChitoPEITC nanophotosensitizers showed significantly higher PDT efficacy against HCT-116 cells than that of free Ce6. These results indicate that ChitoPEITC nanophotosensitizers have superior potential in Ce6 uptake, ROS production and PDT efficacy. In the HCT-116 cell-bearing mice tumor-xenograft model, ChitoPEITC nanophotosensitizers efficiently inhibited growth of tumor volume rather than free Ce6. In the animal imaging study, ChitoPEITC nanophotosensitizers were concentrated in the tumor tissue, i.e., fluorescence intensity in the tumor tissue was stronger than that of other tissues. We suggest that ChitoPEITC nanophotosensitizers are a promising candidate for the treatment of human colon cancer cells.
Collapse
Affiliation(s)
- Inho Bae
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Taeyu Grace Kim
- Tyros Biotechnology Inc., 75 Kneeland St. 14 floors, Boston, MA 02111, USA
- Brookline High School, 115 Greenough St., Brookline, MA 02445, USA
| | - Taeyeon Kim
- College of Art & Science, University of Pennsylvania, 249 S 36th St., Philadelphia, PA 19104, USA
| | - Dohoon Kim
- Tyros Biotechnology Inc., 75 Kneeland St. 14 floors, Boston, MA 02111, USA
| | - Doug-Hoon Kim
- Department of Optometry, Masan University, Changwon 51217, Korea
| | - Jaewon Jo
- Gwangju Center, Korea Basic Science Institute, Gwangju 61186, Korea
| | - Young-Ju Lee
- Gwangju Center, Korea Basic Science Institute, Gwangju 61186, Korea
| | - Young-Il Jeong
- Tyros Biotechnology Inc., 75 Kneeland St. 14 floors, Boston, MA 02111, USA
| |
Collapse
|
19
|
Zhao C, Chen Q, Li W, Zhang J, Yang C, Chen D. Multi-functional platelet membrane-camouflaged nanoparticles reduce neuronal apoptosis and regulate microglial phenotype during ischemic injury. APPLIED MATERIALS TODAY 2022; 27:101412. [DOI: 10.1016/j.apmt.2022.101412] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Chaoyue Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
- Changchun Children's Hospital, 1321Beian Road, Changchun, Jilin 130051, China
| | | | | | | | - Chunrong Yang
- Department of Pharmacy, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| |
Collapse
|
20
|
Ferrari D, Gessi S, Merighi S, Nigro M, Travagli A, Burns JS. Potentiating Cancer Immune Therapy via Nanomaterials and Purinergic Signaling. Front Cell Dev Biol 2022; 10:893709. [PMID: 35602602 PMCID: PMC9114640 DOI: 10.3389/fcell.2022.893709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/28/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Davide Ferrari
- Section of Microbiology and Applied Pathology, Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Stefania Gessi
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Stefania Merighi
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Manuela Nigro
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Alessia Travagli
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Jorge S. Burns
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
21
|
Guo J, Zeng H, Shi X, Han T, Liu Y, Liu Y, Liu C, Qu D, Chen Y. A CFH peptide-decorated liposomal oxymatrine inactivates cancer-associated fibroblasts of hepatocellular carcinoma through epithelial–mesenchymal transition reversion. J Nanobiotechnology 2022; 20:114. [PMID: 35248071 PMCID: PMC8898522 DOI: 10.1186/s12951-022-01311-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
AbstractCancer-associated fibroblasts (CAFs) deteriorate tumor microenvironment (TME) and hinder intra-tumoral drug delivery. Direct depleting CAFs exists unpredictable risks of tumor metastasis. Epithelial–mesenchymal transition (EMT) is a critical process of CAFs converted from hepatic stellate cells during hepatocellular tumorigenesis; however, until now the feasibility of reversing EMT to battle hepatocellular carcinoma has not been comprehensively explored. In this study, we report a CFH peptide (CFHKHKSPALSPVGGG)-decorated liposomal oxymatrine (CFH/OM-L) with a high affinity to Tenascin-C for targeted inactivating CAFs through reversing EMT, which is verified by the upregulation of E-cadherin and downregulation of vimentin, N-cadherin, and snail protein in vivo and in vitro. After the combination with icaritin-loaded lipid complex, CFH/OM-L obviously boosts the comprehensive anticancer efficacy in both 3D tumor spheroids and stromal-rich tumor xenograft nude mouse models. The combinational therapy not only effectively reversed the in vivo EMT process but also significantly lowered the collagen, creating favorable conditions for deep penetration of nanoparticles. More importantly, CFH/OM-L does not kill but inactivates CAFs, resulting in not only a low risk of tumor metastasis but also a reprogramming TME, such as M1 tumor-associated macrophages polarization and natural killer cells activation. Such strategy paves a moderate way to remold TME without depleting CAFs and provides a powerful tool to design strategies of combinational hepatocellular carcinoma therapy.
Graphical Abstract
Collapse
|
22
|
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
|