1
|
Rodponthukwaji K, Khowawisetsut L, Limjunyawong N, Kunwong N, Duangchan K, Sripinitchai S, Sathornsumetee S, Nguyen T, Srisawat C, Punnakitikashem P. Enhanced Anticancer Effects Through Combined Therapeutic Model of Macrophage Polarization and Cancer Cell Apoptosis by Multifunctional Lipid Nanocomposites. J Biomed Mater Res A 2025; 113:e37886. [PMID: 39972623 DOI: 10.1002/jbm.a.37886] [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: 09/28/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
Although the mono-anticancer therapy approach particularly directly targeting tumors is still common, this conventional method is generally deemed not effective and insufficient. In tumor microenvironment (TME), tumor-associated macrophages (TAMs, referred to as M2-polarized) play a crucial role in creating an immunosuppressive TME, contributing to various pro-tumorigenic effects. A promising strategy to inhibit tumor growth involves re-educating M2 macrophages into tumoricidal macrophages (M1). Therefore, combining macrophage reprogramming with cancer cell death induction in a single modality may offer synergistic benefits in cancer therapy. Here, we engineered a lipid-based delivery platform capable of co-delivering resiquimod (R848) and polyinosinic: polycytidylic acid (PIC). R848 in our nanosystem effectively triggered M2-to-M1 repolarization, as evidenced by the upregulation of M1 marker genes (TNF, IL6), the release of proinflammatory cytokines (TNF-α and IL-6), and the downregulation of the M2 marker gene, MRC1. On the other hand, the presence of PIC increased caspase-3/7 activity leading to cancer cell death through the apoptotic pathway. This nanocarrier system established a multifunctional platform to enhance the anticancer effect. The synergistic effect of repolarized macrophages in combination with the induction of apoptosis, facilitated by our nanomedicine, was evident in a co-culture system of macrophage and cancer cells, showing a significant increase in cancer cell death compared to individual treatments. These findings attractively demonstrated the potential of our multifunctional lipid nanoparticles as therapeutic agents for anticancer treatment by modulating the tumor immune microenvironment and simultaneously increasing cancer cell cytotoxicity.
Collapse
Affiliation(s)
- Kamonlatth Rodponthukwaji
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathachit Limjunyawong
- Siriraj Center of Research Excellence in Allergy and Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Natsuda Kunwong
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kongpop Duangchan
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sirinapa Sripinitchai
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sith Sathornsumetee
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tam Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Yilun W, Yaojing Z, Hongcan S. Nanoparticle trends and hotspots in lung cancer diagnosis from 2006-2023: a bibliometric analysis. Front Oncol 2024; 14:1453021. [PMID: 39759141 PMCID: PMC11695240 DOI: 10.3389/fonc.2024.1453021] [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: 06/22/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Background Lung cancer possesses the highest incidence and mortality rates among malignancies globally. Despite substantial advancements in oncology, it is frequently diagnosed at an advanced stage, resulting in a poor prognosis. Over recent decades, the swift progress of nanotechnology has precipitated the extensive utilization of nanomaterials as carriers in cancer diagnosis and therapy. The deployment of nanoparticles as an innovative diagnostic strategy aspires to enable the earlier detection of lung cancer, thereby permitting earlier intervention and enhancing prognosis. This study endeavors to deepen our understanding of this domain through a comprehensive analysis employing bibliometric tools. Method Related articles were retrieved from the Web of Science Core Collection from January 1st, 2006, to December 14st, 2023. Thereaf CiteSpace, VOSviewer and the online platform of bibliometrics (http://bibliometric.com/) were utilized to visually analyze Author/Country/Institutions/Cited Journals/Keyword, et al. Results A total of 966 articles were retrieved for this study. The analysis unveils a progressive increase in annual publications within this field, with China at the forefront in publication volume, followed by the United States and India. Moreover, Chinese research institutions, notably the Chinese Academy of Sciences and Shanghai Jiao Tong University, prevail in publication output. Upon exclusion of irrelevant search terms, keywords clustering analysis highlights that "biomarkers", "sensors", "gold nanoparticles", and "silver nanoparticles" are predominant research focuses. Conclusion This bibliometric study furnishes a quantitative perspective on the extant literature, serving scholars in related fields. Furthermore, it anticipates future research trend concerning nanoparticles and lung cancer diagnosis, thereby aiding in the formulation of project planning and the design of experiments.
Collapse
Affiliation(s)
- Wang Yilun
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhang Yaojing
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shi Hongcan
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Thoracic and Cardiovascular Surgery, Northern Jiangsu Peoples Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
3
|
Park HB, An EK, Kim SJ, Ryu D, Zhang W, Pack CG, Kim H, Kwak M, Im W, Ryu JH, Lee PCW, Jin JO. Anti-PD-L1 Antibody Fragment Linked to Tumor-Targeting Lipid Nanoparticle Can Eliminate Cancer and Its Metastasis via Photoimmunotherapy. ACS NANO 2024; 18:33366-33380. [PMID: 39603816 DOI: 10.1021/acsnano.4c08448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Effective cancer therapy aims to treat primary tumors and metastatic and recurrent cancer. Immune checkpoint blockade-mediated immunotherapy has shown promising effects against tumors; however, its efficacy in metastatic or recurrent cancer is limited. Here, based on the advantages of nanomedicine, lipid nanoparticles (LNPs) that can target tumors are synthesized for photothermal therapy (PTT) and immunotherapy to treat primary and metastatic recurrent cancer. These LNPs, termed piLNPs, are encapsulated with indocyanine green and incorporated with the antigen (Ag)-binding fragment of the anti-PD-L1 antibody for targeting tumors and immunotherapy. Intravenously injected piLNPs in 4T1 breast tumor-bearing BALB/c mice effectively target the 4T1 tumor and are suitable for performing PTT using a near-infrared laser. Moreover, lung metastatic 4T1 tumor growth is completely prevented in mice previously cured of the 4T1 breast tumor by piLNP treatment and rechallenged with lung 4T1 metastatic cancer. Blockage of the second challenged metastatic 4T1 breast cancer by piLNP is due to the activation of Ag-specific T cells. Cytotoxic T lymphocytes from piLNP-cured mice selectively attack 4T1 breast cancer cells. Therefore, piLNP can be used as a multifunctional breast cancer treatment composition that can target tumors, treat primary tumors, and prevent metastasis and recurrence.
Collapse
Affiliation(s)
- Hae-Bin Park
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Chan-Gi Pack
- Department of Biomedical Engineering, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, South Korea
| | - Minseok Kwak
- Department of Chemistry and Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, South Korea
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| |
Collapse
|
4
|
Brüßeler MT, Zam A, Moreno-Zafra VM, Rouatbi N, Hassuneh OWM, Marrocu A, Liam-Or R, Abdel-Bar HM, Walters AA, Al-Jamal KT. Polyinosinic/Polycytidylic Lipid Nanoparticles Enhance Immune Cell Infiltration and Improve Survival in the Glioblastoma Mouse Model. Mol Pharm 2024; 21:6339-6352. [PMID: 39556101 PMCID: PMC11615939 DOI: 10.1021/acs.molpharmaceut.4c00875] [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: 08/04/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024]
Abstract
Glioblastoma (GBM) immunotherapy is particularly challenging due to the pro-tumorigenic microenvironment, marked by low levels and inactive immune cells. Toll-like receptor (TLR) agonists have emerged as potent immune adjuvants but failed to show improved outcomes in clinical trials when administered as a monotherapy. We hypothesize that a combined nanoparticulate formulation of TLR agonist and immunogenic cell death-inducing drug (doxorubicin) will synergize to induce improved GBM immunotherapy. Lipid nanoparticle (LNP) formulations of the TLR agonists CpG and polyinosinic/polycytidylic (pIpC), with and without Dox, were first prepared, achieving an encapsulation efficiency >75% and a size <140 nm. In vitro studies identified that LNP pIpC was superior to CpG at activating bone marrow-derived immune cell populations (dendritic cells and macrophages) with minimal toxicity. It was also observed that the pIpC formulation can skew macrophage polarization toward the antitumorigenic M1 phenotype and increase macrophage phagocytosis of cancer cells. Upon intratumoral administration, pIpC Dox LNPs led to significant immune cell infiltration and activation. In survival models, the inclusion of Dox into pIpC LNP improved mice survival compared to control. However, addition of Dox did not show significant improvement in mice's survival compared to singly formulated pIpC LNP. This study has illustrated the potential of pIpC LNP formulations in prospective GBM immunotherapeutic regimes. Future studies will focus on optimizing dosage regimen and/or combination with other modalities, including the standard of care (temozolomide), immune checkpoint blockade, or cancer vaccines.
Collapse
Affiliation(s)
- Melanie
M. T. Brüßeler
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Ludwig
Maximilians University, Bayern, Munich, München 80539, Germany
| | - Alaa Zam
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Víctor M. Moreno-Zafra
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Nadia Rouatbi
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Osama W. M. Hassuneh
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Alessia Marrocu
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Revadee Liam-Or
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong 999077, China
| | - Hend Mohamed Abdel-Bar
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Department
of Pharmaceutics, Faculty of Pharmacy, University
of Sadat City, P.O. Box 32958, El Sadat, Egypt
| | - Adam Alexander Walters
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
| | - Khuloud T. Al-Jamal
- Institute
of Pharmaceutical Science, King’s
College London, Franklin-Wilkins Building 150 Stamford Street, London SE1 9NH, U.K.
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong 999077, China
| |
Collapse
|
5
|
Chaudhary AA, Fareed M, Khan SUD, Alneghery LM, Aslam M, Alex A, Rizwanullah M. Exploring the therapeutic potential of lipid-based nanoparticles in the management of oral squamous cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1223-1246. [PMID: 39465011 PMCID: PMC11502080 DOI: 10.37349/etat.2024.00272] [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: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a highly malignant and invasive tumor with significant mortality and morbidity. Current treatment modalities such as surgery, radiotherapy, and chemotherapy encounter significant limitations, such as poor targeting, systemic toxicity, and drug resistance. There is an urgent need for novel therapeutic strategies that offer targeted delivery, enhanced efficacy, and reduced side effects. The advent of lipid-based nanoparticles (LNPs) offers a promising tool for OSCC therapy, potentially overcoming the limitations of current therapeutic approaches. LNPs are composed of biodegradable and biocompatible lipids, which minimize the risk of toxicity and adverse effects. LNPs can encapsulate hydrophobic drugs, improving their solubility and stability in the biological environment, thereby enhancing their bioavailability. LNPs demonstrate significantly higher ability to encapsulate lipophilic drugs than other nanoparticle types. LNPs offer excellent storage stability, minimal drug leakage, and controlled drug release, making them highly effective nanoplatforms for the delivery of chemotherapeutic agents. Additionally, LNPs can be modified by complexing them with specific target ligands on their surface. This surface modification allows the active targeting of LNPs to the tumors in addition to the passive targeting mechanism. Furthermore, the PEGylation of LNPs improves their hydrophilicity and enhances their biological half-life by reducing clearance by the reticuloendothelial system. This review aims to discuss current treatment approaches and their limitations, as well as recent advancements in LNPs for better management of OSCC.
Collapse
Affiliation(s)
- Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Fareed
- College of Medicine, AlMaarefa University, Diriyah, Riyadh 11597, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001, Kurdistan Region, Iraq
| | - Arockia Alex
- Molecular and Nanobiotechnology Laboratory (MNBL), Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Md Rizwanullah
- Drug Delivery and Nanomedicine Unit, Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| |
Collapse
|
6
|
Wang J, Guo B, Sun Z, Zhao S, Cao L, Zhong Z, Meng F. Polymersomal Poly(I:C) Self-Magnifies Antitumor Immunity by Inducing Immunogenic Cell Death and Systemic Immune Activation. Adv Healthc Mater 2024; 13:e2400784. [PMID: 38896790 DOI: 10.1002/adhm.202400784] [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: 02/28/2024] [Revised: 06/18/2024] [Indexed: 06/21/2024]
Abstract
Immunotherapy has emerged as a powerful weapon against lung cancer, yet only a fraction of patients respond to the treatment. Poly(I:C) (PIC) effectively triggers both innate and adaptive immunity. It can also induce immunogenic cell death (ICD) in tumor cells. However, its efficacy is hindered by its instability in vivo and limited cellular uptake. To address this, PIC is encapsulated in cRGD-functionalized polymersomes (t-PPIC), which significantly increases its stability and uptake, thus activating dendritic cells (DCs) and inducing apoptosis of lung tumor cells in vitro. In a murine LLC lung tumor model, systemic administration of t-PPIC effectively suppresses tumor growth and leads to survival benefits, with 40% of the mice becoming tumor-free. Notably, t-PPIC provokes stronger apoptosis and ICD in tumor tissue and elicits a more potent stimulation of DCs, recruitment of natural killer (NK) cells, and activation of CD8+ T cells, compared to free PIC and nontargeted PPIC controls. Furthermore, when combined with immune checkpoint inhibitors or radiotherapy, t-PPIC amplifies the antitumor immune response, resulting in complete regression in 60% of the mice. These compelling findings underscore the potential of integrin-targeted polymersomal PIC to enhance antitumor immunity by simultaneously inducing ICD and systemic immune activation.
Collapse
Affiliation(s)
- Jingyi Wang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Zhiwei Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Li Cao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, P. R. China
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215006, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| |
Collapse
|
7
|
Jo S, Roh S, Shim J, Yu JW, Jung Y, Jang WY, Seo B, Won YY, Yoo J. Modulating the Thermoresponsive Characteristics of PLGA-PEG-PLGA Hydrogels via Manipulation of PLGA Monomer Sequences. Biomacromolecules 2024; 25:5374-5386. [PMID: 39014545 DOI: 10.1021/acs.biomac.4c00817] [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: 07/18/2024]
Abstract
Hydrogels are promising materials for biomedical applications, particularly in drug delivery and tissue engineering. This study highlights thermoresponsive hydrogels, specifically poly(lactic-co-glycolic acid) (PLGA)-poly(ethylene glycol) (PEG)-PLGA triblock copolymers, and introduces a feed rate-controlled polymerization (FRCP) method. By utilizing an organic catalyst and regulating the monomer feed rate, the sequence distribution of PLGA within the triblock copolymer is controlled. Various analyses, including 13C NMR and rheological measurements, were conducted to investigate the impact of sequence distribution. Results show that altering sequence distribution significantly influences the sol-gel transition, hydrophobicity-hydrophilicity balance, and drug release profile. Increased sequence uniformity lowers the glass transition temperature, raises the sol-gel transition temperature due to enhanced hydrophilicity, and promotes a more uniform drug (curcumin) distribution within the PLGA domain, resulting in a slower release rate. This study emphasizes the importance of PLGA sequence distribution in biomedical applications and the potential of FRCP to tailor thermoresponsive hydrogels for biomedical advancements.
Collapse
Affiliation(s)
- SeongHoon Jo
- Center of Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Soonjong Roh
- Center of Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaemin Shim
- Department of Chemical Biological Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Ji Woong Yu
- Center for AI and Natural Sciences, Korea Institute for Advanced Study, Seoul 02455, Republic of Korea
| | - Youngmee Jung
- Center of Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Institute of Nano, Regeneration, Reconstruction, Korea University, Seoul 02841, Republic of Korea
| | - Bumjoon Seo
- Department of Chemical Biological Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47906, United States
| | - Jin Yoo
- Center of Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| |
Collapse
|
8
|
Wei M, Wang X, Mo Y, Kong C, Zhang M, Qiu G, Tang Z, Chen J, Wu F. Combined Effects of Anti-PD-L1 and Nanosonodynamic Therapy on HCC Immune Activation in Mice: An Investigation. Int J Nanomedicine 2024; 19:7215-7236. [PMID: 39050875 PMCID: PMC11268760 DOI: 10.2147/ijn.s427144] [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: 06/22/2023] [Accepted: 05/29/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Current therapeutic strategies, including immune checkpoint blockade (ICB), exhibit limited efficacy in treating hepatocellular carcinoma (HCC). Nanoparticles, particularly those that can accumulate specifically within tumors and be activated by sonodynamic therapy (SDT), can induce immunogenic cell death (ICD); however, ICD alone has not achieved satisfactory therapeutic effectiveness. This study investigates whether combining ICB with ICD induced by nanoparticle-mediated SDT could enhance anti-tumor immunity and inhibit HCC growth. Methods We developed an iron-based micelle nanodelivery system encapsulating the Near-Infrared Dye IR-780, which was surface-modified with a cyclic tripeptide composed of arginine-glycine-aspartic acid (cRGD). This led to the synthesis of targeted IR780@FOM-cRGD nanoparticles. These nanoparticles were specifically engineered to kill tumor cells under sonication, activate immunogenic cell death (ICD), and be used in conjunction with immune checkpoint blockade (ICB) for the treatment of hepatocellular carcinoma (HCC). Results The synthesized IR780@FOM-cRGD nanoparticles had an average diameter of 28.23±1.750 nm and a Zeta potential of -23.95±1.926. Confocal microscopy demonstrated that IR780@FOM-cRGD could target HCC cells while minimizing toxicity to healthy cells. Upon sonodynamic activation, these nanoparticles consumed significant amounts of oxygen and generated substantial reactive oxygen species (ROS), effectively killing tumor cells and inhibiting the proliferation, invasion, and migration of H22 cells. Hemolysis assays confirmed the in vivo safety of the nanoparticles, and in vivo fluorescence imaging revealed significant accumulation in tumor tissues. Mouse model experiments showed that combining ICB(which induced by Anti-PD-L1) with ICD (which induced by IR780@FOM-cRGD), could effectively activated anti-tumor immunity and suppressed tumor growth. Discussion This study highlights the potential of IR780@FOM-cRGD nanoparticles to facilitate tumor eradication and immune activation when used in conjunction with Anti-PD-L1 therapy. This combination represents a non-invasive, efficient, and targeted approach for the treatment of hepatocellular carcinoma (HCC). By integrating sonodynamic therapy with immunotherapy, this strategy promises to substantially improve the effectiveness of traditional treatments in combating HCC, offering new avenues for clinical application and therapeutic innovation.
Collapse
Affiliation(s)
- Meng Wei
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Xiaobo Wang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Yunhai Mo
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Cunqing Kong
- Medical Imaging Center, Affiliated Taihe Hospital, Hubei University of Medicine, Hubei, 442000, People’s Republic of China
| | - Mengqi Zhang
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
- Department of Interventional Therapy, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Guanhua Qiu
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
- Department of Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Zhihong Tang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Jie Chen
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Feixiang Wu
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| |
Collapse
|
9
|
Zhang X, Ma Y, Shi Y, Jiang L, Wang L, Ur Rashid H, Yuan M, Liu X. Advances in liposomes loaded with photoresponse materials for cancer therapy. Biomed Pharmacother 2024; 174:116586. [PMID: 38626516 DOI: 10.1016/j.biopha.2024.116586] [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: 02/22/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024] Open
Abstract
Cancer treatment is presently a significant challenge in the medical domain, wherein the primary modalities of intervention include chemotherapy, radiation therapy and surgery. However, these therapeutic modalities carry side effects. Photothermal therapy (PTT) and photodynamic therapy (PDT) have emerged as promising modalities for the treatment of tumors in recent years. Phototherapy is a therapeutic approach that involves the exposure of materials to specific wavelengths of light, which can subsequently be converted into either heat or Reactive Oxygen Species (ROS) to effectively eradicate cancer cells. Due to the hydrophobicity and lack of targeting of many photoresponsive materials, the use of nano-carriers for their transportation has been extensively explored. Among these nanocarriers, liposomes have been identified as an effective drug delivery system due to their controllability and availability in the biomedical field. By binding photoresponsive materials to liposomes, it is possible to reduce the cytotoxicity of the material and regulate drug release and accumulation at the tumor site. This article provides a comprehensive review of the progress made in cancer therapy using photoresponsive materials loaded onto liposomes. Additionally, the article discusses the potential synergistic treatment through the combination of phototherapy with chemo/immuno/gene therapy using liposomes.
Collapse
Affiliation(s)
- Xianwei Zhang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Youfu Ma
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Yenong Shi
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Lihe Jiang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Lisheng Wang
- Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Pelotas, RS 96010-900, Brazil
| | - Haroon Ur Rashid
- Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Pelotas, RS 96010-900, Brazil
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| |
Collapse
|
10
|
Paun RA, Jurchuk S, Tabrizian M. A landscape of recent advances in lipid nanoparticles and their translational potential for the treatment of solid tumors. Bioeng Transl Med 2024; 9:e10601. [PMID: 38435821 PMCID: PMC10905562 DOI: 10.1002/btm2.10601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 03/05/2024] Open
Abstract
Lipid nanoparticles (LNPs) are biocompatible drug delivery systems that have found numerous applications in medicine. Their versatile nature enables the encapsulation and targeting of various types of medically relevant molecular cargo, including oligonucleotides, proteins, and small molecules for the treatment of diseases, such as cancer. Cancers that form solid tumors are particularly relevant for LNP-based therapeutics due to the enhanced permeation and retention effect that allows nanoparticles to accumulate within the tumor tissue. Additionally, LNPs can be formulated for both locoregional and systemic delivery depending on the tumor type and stage. To date, LNPs have been used extensively in the clinic to reduce systemic toxicity and improve outcomes in cancer patients by encapsulating chemotherapeutic drugs. Next-generation lipid nanoparticles are currently being developed to expand their use in gene therapy and immunotherapy, as well as to enable the co-encapsulation of multiple drugs in a single system. Other developments include the design of targeted LNPs to specific cells and tissues, and triggerable release systems to control cargo delivery at the tumor site. This review paper highlights recent developments in LNP drug delivery formulations and focuses on the treatment of solid tumors, while also discussing some of their current translational limitations and potential opportunities in the field.
Collapse
Affiliation(s)
- Radu A. Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Sarah Jurchuk
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Faculty of Dentistry and Oral Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
11
|
Lee C. Albumin hydrogels for repeated capture of drugs from the bloodstream and release into the tumor. J Control Release 2024; 365:384-397. [PMID: 38007193 DOI: 10.1016/j.jconrel.2023.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Despite the efficacy of hydrogels for consistently delivering drugs to targeted areas (primarily tumors), these systems face challenges such as initial burst release, non-refillable drugs, and a lack of dosage control. To address these issues, a novel strategy has been developed to capture and release drugs from the bloodstream, thereby overcoming the limitations of traditional hydrogels. In this study, an innovative albumin hydrogel system was developed through a bioorthogonal reaction using azide-modified albumin and 4-arm PEG-DBCO. This system can repeatedly capture and release drugs over prolonged periods. Inspired by albumin-drug binding in vivo, this hydrogel can be injected intratumorally and acts as a reservoir for capturing drugs circulating in the bloodstream. Drugs captured in hydrogels are released slowly and effectively delivered to tumors through a "capture and release process." Both the in vitro and in vivo results indicated that the hydrogel effectively captured and released drugs, such as indocyanine green and doxorubicin, over repeated cycles without compromising the activity of the drugs. Moreover, implanting the hydrogel at surgical sites successfully inhibited tumor recurrence through its drug capture-release capability. These findings establish the albumin hydrogel system as a promising capture-release platform that leverages drug-binding affinity to effectively deliver drugs to tumors, offering potential advancements in cancer treatment and post-surgery recurrence prevention.
Collapse
Affiliation(s)
- Changkyu Lee
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 38066, Republic of Korea.
| |
Collapse
|
12
|
Youness RA, Mohamed AH, Efthimiadou EK, Mekky RY, Braoudaki M, Fahmy SA. A Snapshot of Photoresponsive Liposomes in Cancer Chemotherapy and Immunotherapy: Opportunities and Challenges. ACS OMEGA 2023; 8:44424-44436. [PMID: 38046305 PMCID: PMC10688172 DOI: 10.1021/acsomega.3c04134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/18/2023] [Indexed: 12/05/2023]
Abstract
To provide precise medical regimens, photonics technologies have been involved in the field of nanomedicine. Phototriggered liposomes have been cast as promising nanosystems that achieve controlled release of payloads in several pathological conditions such as cancer, autoimmune, and infectious diseases. In contrast to the conventional liposomes, this photoresponsive element greatly improves therapeutic efficacy and reduces the adverse effects of gene/drug therapy during treatment. Recently, cancer immunotherpay has been one of the hot topics in the field of oncology due to the great success and therapeutic benefits that were well-recognized by the patients. However, several side effects have been encountered due to the unmonitored augmentation of the immune system. This Review highlights the most recent advancements in the development of photoresponsive liposome nanosystems in the field of oncology, with a specific emphasis on challenges and opportunities in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Rana A. Youness
- Biology
and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, Cairo 4824201, Egrypt
- Biology
and Biochemistry Department, Molecular Genetics Research Team (MGRT),
School of Life and Medical Sciences, University
of Hertfordshire Hosted by Global Academic Foundation, Cairo 11835, Egypt
| | - Adham H. Mohamed
- Department
of Chemistry, Faculty of Science, Cairo
University, Giza 12613, Egypt
| | - Eleni K. Efthimiadou
- Inorganic
Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou 157 71, Greece
| | - Radwa Y. Mekky
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
| | - Maria Braoudaki
- Clinical,
Pharmaceutical, and Biological Science Department, School of Life
and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, U.K.
| | - Sherif Ashraf Fahmy
- Chemistry
Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo 11835, Egypt
| |
Collapse
|
13
|
Min SH, Lei W, Jun CJ, Yan ZS, Guang YX, Tong Z, Yong ZP, Hui LZ, Xing H. Design strategy and research progress of multifunctional nanoparticles in lung cancer therapy. Expert Opin Investig Drugs 2023; 32:723-739. [PMID: 37668152 DOI: 10.1080/13543784.2023.2254683] [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: 04/25/2023] [Revised: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
INTRODUCTION Lung cancer is one of the cancer types with the highest mortality rate, exploring a more effective treatment modality that improves therapeutic efficacy while mitigating side effects is now an urgent requirement. Designing multifunctional nanoparticles can be used to overcome the limitations of drugs and conventional drug delivery systems. Nanotechnology has been widely researched, and through different needs, suitable nanocarriers can be selected to load anti-cancer drugs to improve the therapeutic effect. It is foreseeable that with the rapid development of nanotechnology, more and more lung cancer patients will benefit from nanotechnology. This paper reviews the merits of various multifunctional nanoparticles in the treatment of lung cancer to provide novel ideas for lung cancer treatment. AREAS COVERED This review focuses on summarizing various nanoparticles for targeted lung cancer therapy and their advantages and disadvantages, using nanoparticles loaded with anti-cancer drugs, delivered to lung cancer sites, enhancing drug half-life, improving anti-cancer drug efficacy and reducing side effects. EXPERT OPINION The delivery mode of nanoparticles with superior pharmacokinetic properties in the in vivo circulation enhances the half-life of the drug, and provides tissue-targeted selectivity and the ability to overcome biological barriers, bringing a revolution in the field of oncology.
Collapse
Affiliation(s)
- Shen Hui Min
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Lei
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Jia Jun
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhang Shao Yan
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Xu Guang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhang Tong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Pei Yong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Zhen Hui
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huang Xing
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Kansız S, Elçin YM. Advanced liposome and polymersome-based drug delivery systems: Considerations for physicochemical properties, targeting strategies and stimuli-sensitive approaches. Adv Colloid Interface Sci 2023; 317:102930. [PMID: 37290380 DOI: 10.1016/j.cis.2023.102930] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023]
Abstract
Liposomes and polymersomes are colloidal vesicles that are self-assembled from lipids and amphiphilic polymers, respectively. Because of their ability to encapsulate both hydrophilic and hydrophobic therapeutics, they are of great interest in drug delivery research. Today, the applications of liposomes and polymersomes have expanded to a wide variety of complex therapeutic molecules, including nucleic acids, proteins and enzymes. Thanks to their chemical versatility, they can be tailored to different drug delivery applications to achieve maximum therapeutic index. This review article evaluates liposomes and polymersomes from a perspective that takes into account the physical and biological barriers that reduce the efficiency of the drug delivery process. In this context, the design approaches of liposomes and polymersomes are discussed with representative examples in terms of their physicochemical properties (size, shape, charge, mechanical), targeting strategies (passive and active) and response to different stimuli (pH, redox, enzyme, temperature, light, magnetic field, ultrasound). Finally, the challenges limiting the transition from laboratory to practice, recent clinical developments, and future perspectives are addressed.
Collapse
Affiliation(s)
- Seyithan Kansız
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey; Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
15
|
Zhang JY, Gao WD, Lin JY, Xu S, Zhang LJ, Lu XC, Luan X, Peng JQ, Chen Y. Nanotechnology-based photo-immunotherapy: a new hope for inhibition of melanoma growth and metastasis. J Drug Target 2023:1-14. [PMID: 37216425 DOI: 10.1080/1061186x.2023.2216402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023]
Abstract
Melanoma is the most aggressive form of skin cancer and there is a need for the development of effective anti-melanoma therapies as it shows high metastatic ability and low response rate. In addition, it has been identified that traditional phototherapy could trigger immunogenic cell death (ICD) to activate antitumor immune response, which could not only effectively arrest primary tumor growth, but also exhibit superior effects in terms of anti-metastasis, anti-recurrence for metastatic melanoma treatment However, the limited tumor accumulation of photosensitizers/photothermal agents and immunosuppressive tumor microenvironment severely weaken the immune effects. The application of nanotechnology facilitates a higher accumulation of photosensitizers/photothermal agents at the tumor site, which can thus improve the antitumor effects of photo-immunotherapy (PIT). In this review, we summarize the basic principles of nanotechnology-based PIT and highlight novel nanotechnologies that are expected to enhance the antitumor immune response for improved therapeutic efficacy.
Collapse
Affiliation(s)
- Ji-Yuan Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Dong Gao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Yi Lin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin-Chen Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian-Qing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
16
|
Yadav D, Puranik N, Meshram A, Chavda V, Lee PCW, Jin JO. How Advanced are Cancer Immuno-Nanotherapeutics? A Comprehensive Review of the Literature. Int J Nanomedicine 2023; 18:35-48. [PMID: 36636642 PMCID: PMC9830082 DOI: 10.2147/ijn.s388349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
Cancer is a broad term for a group of diseases involving uncontrolled cell growth and proliferation. There is no cure for cancer despite recent significant improvements in screening, treatment, and prevention approaches. Among the available treatments, immunotherapy has been successful in targeting and killing cancer cells by stimulating or enhancing the body's immune system. Antibody-based immunotherapeutic agents that block immune checkpoint proteins expressed by cancer cells have shown promising results. The rapid development of nanotechnology has contributed to improving the effectiveness and reducing the adverse effects of these anti-cancer immunotherapeutic agents. Recently, engineered nanomaterials have been the focus of many state-of-The-art approaches toward effective cancer treatment. In this review, the contribution of various nanomaterials such as polymeric nanoparticles, dendrimers, microspheres, and carbon nanomaterials in improving the efficiency of anti-cancer immunotherapy is discussed as well as nanostructures applied to combination cancer immunotherapy.
Collapse
Affiliation(s)
- Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Nidhi Puranik
- Biological Sciences Department, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Anju Meshram
- Department of Biotechnology, Kalinga University, Naya Raipur, Chhattisgarh, India
| | - Vishal Chavda
- Department of Pathology, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Peter Chang-Whan Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, South Korea,Correspondence: Peter Chang-Whan Lee, Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, South Korea, Email
| | - Jun-O Jin
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, 05505, South Korea,Jun-O Jin, Department of Microbiology, University of Ulsan College of Medicine, Seoul, 05505, South Korea, Email
| |
Collapse
|
17
|
Biomaterial-assisted photoimmunotherapy for synergistic suppression of cancer progression. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
18
|
Dinakar YH, Karole A, Parvez S, Jain V, Mudavath SL. Organ-restricted delivery through stimuli-responsive nanocarriers for lung cancer therapy. Life Sci 2022; 310:121133. [DOI: 10.1016/j.lfs.2022.121133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
|
19
|
Gao Q, Lee JS, Kim BS, Gao G. Three-dimensional printing of smart constructs using stimuli-responsive biomaterials: A future direction of precision medicine. Int J Bioprint 2022; 9:638. [PMID: 36636137 PMCID: PMC9830998 DOI: 10.18063/ijb.v9i1.638] [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: 06/23/2022] [Accepted: 09/10/2022] [Indexed: 11/10/2022] Open
Abstract
Three-dimensional (3D) printing, which is a valuable technique for the fabrication of tissue-engineered constructs and biomedical devices with complex architectures, has brought about considerable progress in regenerative medicine, drug delivery, and diagnosis of diseases. However, because of the static and inanimate properties of conventional 3D-printed structures, it is difficult to use them in therapies for active and precise medicine, such as improved tissue regeneration, targeted or controlled drug delivery, and advanced pathophysiological monitoring. The integration of stimuli-responsive biomaterials into 3D printing provides a potential strategy for designing and building smart constructs that exhibit programmed functions and controllable changes in properties in response to exogenous and autogenous stimuli. These features make 3D-printed smart constructs the next generation of tissue-engineered products. In this review, we introduce the prevalent 3D printing techniques (with an emphasis on the differences between 3D printing and bioprinting, and biomaterials and bioink), the working principle of each technique, and the advantages of using 3D printing for the fabrication of smart constructs. Stimuli-responsive biomaterials that are widely used for 3D printing of smart constructs are categorized, followed by a summary of their applications in tissue regeneration, drug delivery, and biosensors. Finally, the challenges and future perspectives of 3D-printed smart constructs are discussed.
Collapse
Affiliation(s)
- Qiqi Gao
- School of Medical Engineering, Beijing Institute of Technology, Beijing 100081, China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| | - Jae-Seong Lee
- Department of Information Convergence Engineering, Pusan National University, Yangsan 50612, South Korea
| | - Byoung Soo Kim
- Department of Information Convergence Engineering, Pusan National University, Yangsan 50612, South Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, South Korea
| | - Ge Gao
- School of Medical Engineering, Beijing Institute of Technology, Beijing 100081, China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
20
|
Gamage RS, Smith BD. Spontaneous Transfer of Indocyanine Green from Liposomes to Albumin Is Inhibited by the Antioxidant α-Tocopherol. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:11950-11961. [PMID: 36126324 PMCID: PMC9897306 DOI: 10.1021/acs.langmuir.2c01715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Indocyanine Green (ICG) is a clinically approved organic dye with near-infrared absorption and fluorescence. Over the years, many efforts to improve the photophysical and pharmacokinetic properties of ICG have investigated numerous nanoparticle formulations, especially liposomes with membrane-embedded ICG. A series of systematic absorption and fluorescence experiments, including FRET experiments using ICG as a fluorescence energy acceptor, found that ICG transfers spontaneously from liposomes to albumin protein residing in the external solution with a half-life of ∼10 min at 37 °C. Moreover, transfer of ICG from liposome membranes to external albumin reduces light-activated leakage from thermosensitive liposomes with membrane-embedded ICG. A survey of lipophilic liposome additives discovered that the presence of clinically approved antioxidant, α-tocopherol, greatly increases ICG retention in the liposomes (presumably by forming favorable aromatic stacking interactions), inhibits ICG photobleaching and prevents albumin-induced reduction of light-triggered liposome leakage. This new insight will help researchers with the specific task of optimizing ICG-containing liposomes for fluorescence imaging or phototherapeutics. More broadly, the results suggest a broader design concept concerning light triggered liposome leakage, that is, proximity of the light absorbing dye to the bilayer membrane is a critical design feature that impacts the extent of liposome leakage.
Collapse
|
21
|
Lopes J, Rodrigues CMP, Gaspar MM, Reis CP. Melanoma Management: From Epidemiology to Treatment and Latest Advances. Cancers (Basel) 2022; 14:4652. [PMID: 36230575 PMCID: PMC9562203 DOI: 10.3390/cancers14194652] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is the deadliest skin cancer, whose morbidity and mortality indicators show an increasing trend worldwide. In addition to its great heterogeneity, melanoma has a high metastatic potential, resulting in very limited response to therapies currently available, which were restricted to surgery, radiotherapy and chemotherapy for many years. Advances in knowledge about the pathophysiological mechanisms of the disease have allowed the development of new therapeutic classes, such as immune checkpoint and small molecule kinase inhibitors. However, despite the incontestable progress in the quality of life and survival rates of the patients, effectiveness is still far from desired. Some adverse side effects and resistance mechanisms are the main barriers. Thus, the search for better options has resulted in many clinical trials that are now investigating new drugs and/or combinations. The low water solubility of drugs, low stability and rapid metabolism limit the clinical potential and therapeutic use of some compounds. Thus, the research of nanotechnology-based strategies is being explored as the basis for the broad application of different types of nanosystems in the treatment of melanoma. Future development focus on challenges understanding the mechanisms that make these nanosystems more effective.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, IBEB, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
22
|
Niu Q, Sun Q, Bai R, Zhang Y, Zhuang Z, Zhang X, Xin T, Chen S, Han B. Progress of Nanomaterials-Based Photothermal Therapy for Oral Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:10428. [PMID: 36142341 PMCID: PMC9499573 DOI: 10.3390/ijms231810428] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 12/06/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the top 15 most prevalent cancers worldwide. However, the current treatment models for OSCC (e.g., surgery, chemotherapy, radiotherapy, and combination therapy) present several limitations: damage to adjacent healthy tissue, possible recurrence, low efficiency, and severe side effects. In this context, nanomaterial-based photothermal therapy (PTT) has attracted extensive research attention. This paper reviews the latest progress in the application of biological nanomaterials for PTT in OSCC. We divide photothermal nanomaterials into four categories (noble metal nanomaterials, carbon-based nanomaterials, metal compounds, and organic nanomaterials) and introduce each category in detail. We also mention in detail the drug delivery systems for PTT of OSCC and briefly summarize the applications of hydrogels, liposomes, and micelles. Finally, we note the challenges faced by the clinical application of PTT nanomaterials and the possibility of further improvement, providing direction for the future research of PTT in OSCC treatment.
Collapse
Affiliation(s)
- Qin Niu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Qiannan Sun
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Rushui Bai
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunfan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Zimeng Zhuang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Xin Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianyi Xin
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Si Chen
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Bing Han
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
23
|
How to Treat Melanoma? The Current Status of Innovative Nanotechnological Strategies and the Role of Minimally Invasive Approaches like PTT and PDT. Pharmaceutics 2022; 14:pharmaceutics14091817. [PMID: 36145569 PMCID: PMC9504126 DOI: 10.3390/pharmaceutics14091817] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the most aggressive type of skin cancer, the incidence and mortality of which are increasing worldwide. Its extensive degree of heterogeneity has limited its response to existing therapies. For many years the therapeutic strategies were limited to surgery, radiotherapy, and chemotherapy. Fortunately, advances in knowledge have allowed the development of new therapeutic strategies. Despite the undoubted progress, alternative therapies are still under research. In this context, nanotechnology is also positioned as a strong and promising tool to develop nanosystems that act as drug carriers and/or light absorbents to potentially improve photothermal and photodynamic therapies outcomes. This review describes the latest advances in nanotechnology field in the treatment of melanoma from 2011 to 2022. The challenges in the translation of nanotechnology-based therapies to clinical applications are also discussed. To sum up, great progress has been made in the field of nanotechnology-based therapies, and our understanding in this field has greatly improved. Although few therapies based on nanoparticulate systems have advanced to clinical trials, it is expected that a large number will come into clinical use in the near future. With its high sensitivity, specificity, and multiplexed measurement capacity, it provides great opportunities to improve melanoma treatment, which will ultimately lead to enhanced patient survival rates.
Collapse
|
24
|
Recombinant programmed cell death protein 1 functions as an immune check point blockade and enhances anti-cancer immunity. Biomaterials 2022; 285:121550. [DOI: 10.1016/j.biomaterials.2022.121550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/22/2022] [Accepted: 04/25/2022] [Indexed: 11/21/2022]
|
25
|
Hwang J, An EK, Zhang W, Kim HJ, Eom Y, Jin JO. Dual-functional alginate and collagen–based injectable hydrogel for the treatment of cancer and its metastasis. J Nanobiotechnology 2022; 20:245. [PMID: 35643505 PMCID: PMC9148466 DOI: 10.1186/s12951-022-01458-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/08/2022] [Indexed: 12/19/2022] Open
Abstract
Background Immunotherapies have been gaining attention for the prevention of cancer recurrence and metastasis. Cancer immunotherapy can induce memory cells to target cancer-specific antigens and, thus, selectively kill cancer cells. However, there are difficulties in inducing cancer antigen–specific immunity due to limited knowledge regarding cancer antigens. In this study, we synthesized a dual-functional hydrogel to induce antigen generation and immune activation. Results To elicit a cancer self-antigen–specific immune response, we synthesized an alginate-collagen–based injectable hydrogel, called thermally responsive hydrogel (pTRG), which was incorporated with indocyanine green and the immune stimulator polyinosinic:polycytidylic acid (poly I:C). pTRG was evaluated for its anticancer and anti-metastatic effects against CT-26 carcinoma and 4T1 breast tumor in mice by combining photothermal therapy (PTT) and immunotherapy. Near-infrared (NIR) irradiation promoted temperature elevation in pTRG, consequently exerting a therapeutic effect on mouse tumors. Lung metastasis was prevented in cured CT-26 tumor-injected mice following pTRG treatment via cancer antigen–specific T cell immunity. Moreover, pTRG successfully eliminated the original tumor in 4T1 tumor-bearing mice via PTT and protected them from lung metastasis. To further evaluate the carrier function of TRGs, different types of immunotherapeutic molecules were incorporated into TRGs, which led to the effective elimination of the first CT-26 tumor and the prevention of lung metastasis. Conclusions Our data demonstrate that TRG is a efficient material not only for treating primary tumors but also for preventing metastasis and recurrence.
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01458-x.
Collapse
|
26
|
Hwang J, An EK, Kim SJ, Zhang W, Jin JO. Escherichia coli Mimetic Gold Nanorod-Mediated Photo- and Immunotherapy for Treating Cancer and Its Metastasis. ACS NANO 2022; 16:8472-8483. [PMID: 35466668 DOI: 10.1021/acsnano.2c03379] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Most cancer-related deaths are due to metastasis or recurrence. Therefore, the ultimate goal of cancer therapy will be to treat metastatic and recurrent cancers. Combination therapy for cancer will be one of trial for effective treating metastasis and recurrence. In this study, Escherichia coli-mimetic nanomaterials are synthesized using Escherichia coli membrane proteins, adhesion proteins, and gold nanorods, which are named E. coli mimetic AuNRs (ECA), for combination therapy against cancer and its recurrence. ECA treatment with 808 nm laser irradiation eliminates CT-26 or 4T1 tumors via a photothermal effect. ECA with laser irradiation induces activation of immune cells in the tumor-draining lymph nodes. The mice cured from CT-26 or 4T1 tumor by ECA are rechallenged with those cancer in the lung metastatic form, and the results showed that ECA treatment for the first CT-26 or 4T1 tumor challenge prevents cancer infiltration to the lung in the second challenge. This preventive effect of ECA against tumor growth in the second challenge is aided by cancer antigen-specific T cell immunity. Overall, these findings show that ECA is a nanomaterial with dual functions as a photothermal therapy for treating primary cancers and as immunotherapy for preventing recurrence and metastasis.
Collapse
Affiliation(s)
- Juyoung Hwang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Eun-Koung An
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - So-Jung Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China
| | - Jun-O Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| |
Collapse
|
27
|
Shim MK, Song SK, Jeon SI, Hwang KY, Kim K. Nano-sized drug delivery systems to potentiate the immune checkpoint blockade therapy. Expert Opin Drug Deliv 2022; 19:641-652. [DOI: 10.1080/17425247.2022.2081683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Man Kyu Shim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Su Kyung Song
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Biosystems & Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Seong Ik Jeon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Kwang Yeon Hwang
- Department of Biosystems & Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
28
|
Heptamethine Cyanine-Loaded Nanomaterials for Cancer Immuno-Photothermal/Photodynamic Therapy: A Review. Pharmaceutics 2022; 14:pharmaceutics14051015. [PMID: 35631600 PMCID: PMC9144181 DOI: 10.3390/pharmaceutics14051015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
The development of strategies capable of eliminating metastasized cancer cells and preventing tumor recurrence is an exciting and extremely important area of research. In this regard, therapeutic approaches that explore the synergies between nanomaterial-mediated phototherapies and immunostimulants/immune checkpoint inhibitors have been yielding remarkable results in pre-clinical cancer models. These nanomaterials can accumulate in tumors and trigger, after irradiation of the primary tumor with near infrared light, a localized temperature increase and/or reactive oxygen species. These effects caused damage in cancer cells at the primary site and can also (i) relieve tumor hypoxia, (ii) release tumor-associated antigens and danger-associated molecular patterns, and (iii) induced a pro-inflammatory response. Such events will then synergize with the activity of immunostimulants and immune checkpoint inhibitors, paving the way for strong T cell responses against metastasized cancer cells and the creation of immune memory. Among the different nanomaterials aimed for cancer immuno-phototherapy, those incorporating near infrared-absorbing heptamethine cyanines (Indocyanine Green, IR775, IR780, IR797, IR820) have been showing promising results due to their multifunctionality, safety, and straightforward formulation. In this review, combined approaches based on phototherapies mediated by heptamethine cyanine-loaded nanomaterials and immunostimulants/immune checkpoint inhibitor actions are analyzed, focusing on their ability to modulate the action of the different immune system cells, eliminate metastasized cancer cells, and prevent tumor recurrence.
Collapse
|
29
|
Haegebaert RM, Kempers M, Ceelen W, Lentacker I, Remaut K. Nanoparticle mediated targeting of toll-like receptors to treat colorectal cancer. Eur J Pharm Biopharm 2022; 172:16-30. [PMID: 35074555 DOI: 10.1016/j.ejpb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
30
|
Liu H, Jia D, Yuan F, Wang F, Wei D, Tang X, Tian B, Zheng S, Sun R, Shi J, Fan Q. Her3-specific affibody mediated tumor targeting delivery of ICG enhanced the photothermal therapy against Her3-positive tumors. Int J Pharm 2022; 617:121609. [PMID: 35217073 DOI: 10.1016/j.ijpharm.2022.121609] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/30/2022] [Accepted: 02/19/2022] [Indexed: 02/08/2023]
Abstract
Photothermal therapy (PTT), mediated by tumor-targeted drug delivery of indocyanine green (ICG), is a promising strategy for cancer therapy. Human epidermal growth factor receptor 3 (Her3) is highly expressed in several solid tumors and is an ideal target for tumor diagnosis and therapy. This study prepared a Her3-specific dimeric affibody (ZHer3) using an Escherichia coli expression system. The affibody could bind explicitly to Her3-positive MCF7 and LS174T cells, rather than to Her3-negative SKOV-3 cells in vitro. ICG was coupled with the ZHer3 affibody (ICG-ZHer3) through an N-hydroxysuccinimide (NHS) ester reactive group for tumor-targeted delivery. As expected, Her3-positive cells were selectively and efficiently killed by ICG-ZHer3-mediated PTT in vitro. In vivo, ICG-ZHer3 preferentially accumulated in Her3-positive LS174T tumor grafts because of the tumor-targeting ability of the ZHer3 affibody. As a result of the local generation of cytotoxic reactive oxygen species and hyperthermia, the growth rates of LS174T tumor grafts were significantly inhibited by ICG-ZHer3-mediated PTT, and ICG-ZHer3 showed good safety performance during short-term treatment. In conclusion, these results demonstrated that ICG-ZHer3 is a promising photosensitizer for PTT against Her3-positive tumors.
Collapse
Affiliation(s)
- Huimin Liu
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng 252000, PR China
| | - Feifei Wang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Danfeng Wei
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiaohui Tang
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Baoqing Tian
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Shuhui Zheng
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Ruohan Sun
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Jing Shi
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| |
Collapse
|
31
|
Yadav D, Kwak M, Chauhan PS, Puranik N, Lee PCW, Jin JO. Cancer immunotherapy by immune checkpoint blockade and its advanced application using bio-nanomaterials. Semin Cancer Biol 2022; 86:909-922. [PMID: 35181474 DOI: 10.1016/j.semcancer.2022.02.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 02/07/2023]
Abstract
Cancer is the second leading cause of death worldwide. Traditional approaches, such as surgery, chemotherapy, and radiotherapy have been the main cancer therapeutic modalities in recent years. Cancer immunotherapy is a novel therapeutic modality that potentiates the immune responses of patients against malignancy. Immune checkpoint proteins expressed on T cells or tumor cells serve as a target for inhibiting T cell overactivation, maintaining the balance between self-reactivity and autoimmunity. Tumors essentially hijack the immune checkpoint pathway in order to survive and spread. Immune checkpoint inhibitors (ICIs) are being developed as a result to reactivate the anti-tumor immune response. Recent advances in nanotechnology have contributed to the development of successful, safe, and efficient anticancer drug systems based on nanoparticles. Nanoparticle-based cancer immunotherapy overcomes numerous challenges and offers novel strategies for improving conventional immunotherapies. The fundamental and physiochemical properties of nanoparticles depend on various cancer therapeutic strategies, such as chemotherapeutics, nucleic acid-based treatments, photothermal therapy, and photodynamic agents. The review discusses the use of nanoparticles as carriers for delivering immune checkpoint inhibitors and their efficacy in cancer combination therapy.
Collapse
Affiliation(s)
- Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Minseok Kwak
- Department of Chemistry and Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, South Korea
| | | | - Nidhi Puranik
- Biological Sciences Department, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Peter C W Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul, South Korea.
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| |
Collapse
|
32
|
Zhang L, Jia H, Liu X, Zou Y, Sun J, Liu M, Jia S, Liu N, Li Y, Wang Q. Heptamethine Cyanine–Based Application for Cancer Theranostics. Front Pharmacol 2022; 12:764654. [PMID: 35222006 PMCID: PMC8874131 DOI: 10.3389/fphar.2021.764654] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/09/2021] [Indexed: 01/31/2023] Open
Abstract
Cancer is the most common life-threatening malignant disease. The future of personalized cancer treatments relies on the development of functional agents that have tumor-targeted anticancer activities and can be detected in tumors through imaging. Cyanines, especially heptamethine cyanine (Cy7), have prospective application because of their excellent tumor-targeting capacity, high quantum yield, low tissue autofluorescence, long absorption wavelength, and low background interference. In this review, the application of Cy7 and its derivatives in tumors is comprehensively explored. Cy7 is enormously acknowledged in the field of non-invasive therapy that can “detect” and “kill” tumor cells via near-infrared fluorescence (NIRF) imaging, photothermal therapy (PTT), and photodynamic therapy (PDT). Furthermore, Cy7 is more available and has excellent properties in cancer theranostics by the presence of multifunctional nanoparticles via fulfilling multimodal imaging and combination therapy simultaneously. This review provides a comprehensive scope of Cy7’s application for cancer NIRF imaging, phototherapy, nanoprobe-based combination therapy in recent years. A deeper understanding of the application of imaging and treatment underlying Cy7 in cancer may provide new strategies for drug development based on cyanine. Thus, the review will lead the way to new types with optical properties and practical transformation to clinical practice.
Collapse
Affiliation(s)
- Lei Zhang
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
| | - Hang Jia
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Xuqian Liu
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yaxin Zou
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Jiayi Sun
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Mengyu Liu
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Shuangshuang Jia
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
| | - Nan Liu
- Obstetrics Department, Kaifeng Maternity Hospital, Kaifeng, China
| | - Yanzhang Li
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
- *Correspondence: Qun Wang, ; Yanzhang Li,
| | - Qun Wang
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
- *Correspondence: Qun Wang, ; Yanzhang Li,
| |
Collapse
|
33
|
Lu Y, Wu C, Yang Y, Chen X, Ge F, Wang J, Deng J. Inhibition of tumor recurrence and metastasis via a surgical tumor-derived personalized hydrogel vaccine. Biomater Sci 2022; 10:1352-1363. [PMID: 35112690 DOI: 10.1039/d1bm01596f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tumor recurrence and metastasis have become thorny problems in clinical tumor therapy. Vaccine-mediated antitumor immune response has emerged as a significant postoperative inhibition for tumor recurrence and metastasis. However, limited tumor antigens are not conducive to trigger complete antigen-specific T cell-mediated immune responses. Herein, the design of a hydrogel vaccine system containing a granulocyte-macrophage colony stimulating factor (GM-CSF), based on surgically removed tumor cell lysates, was reported. The hydrogel was formed by crosslinking tumor cell lysates and alginate at low temperatures. The GM-CSF was released from the hydrogel to recruit dendritic cells (DCs), which provided a completely personalized tumor antigen pool. They were combined to foster the production of powerful antigen-specific T cells. The personalized hydrogel was implanted at the surgical site and it stimulated the antitumor immune response for the inhibition of residual tumor cells. Delightfully, the personalized hydrogel inhibited the tumor recurrence and metastasis well in a post-surgical mice tumor model, in combination with a programmed death-ligand 1 antibody (αPD-L1). The results demonstrated that the development of a personalized hydrogel and a combination of αPD-L1 provided a new strategy to prevent tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Yi Lu
- Nanshan School, Guangzhou Medical University, Guangzhou, 510810, China
| | - Chenghu Wu
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China. .,Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.,Oujiang Laboratory, (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Yanyan Yang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xiangzhong Chen
- Multi-Scale Robotics Lab (MSRL), Institute of Robotics and Intelligent System (IRIS) Swiss Federal Institute of Technology (ETH) Zurich, CLA H11.1, ETH-Zentrum, Tannenstrasse 3, CH-8092 Zurich, Switzerland
| | - Feihang Ge
- Hangzhou Chinese Academy of Sciences-Hangzhou Medical College Advanced Medical Technology Institute, Hangzhou 320000, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China. .,Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.,Oujiang Laboratory, (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Junjie Deng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China. .,Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.,Oujiang Laboratory, (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| |
Collapse
|
34
|
Stimuli-responsive nanoliposomes as prospective nanocarriers for targeted drug delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102916] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
35
|
Harnessing the combined potential of cancer immunotherapy and nanomedicine: A new paradigm in cancer treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102492. [PMID: 34775062 DOI: 10.1016/j.nano.2021.102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/16/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022]
Abstract
Cancer immunotherapy has recently emerged as a rising star due to its ability to activate patients' immune systems to fight tumors and prevent relapse. Conversely, the interest in cancer nanomedicine has seemingly waned due to its lackluster clinical translation. Despite being hailed as a game-changer in oncology, cancer immunotherapy still faces numerous challenges. Combining both entities together has thus been one among several solutions proposed to circumvent these challenges. This solution has since gained traction and has also led to a renaissance of cancer nanomedicine. While most combinations are currently experimental at best, some have progressed on to clinical trials. This review thus seeks to examine the advantages and disadvantages of integrating both modalities as a cancer treatment. The opportunities, challenges and future directions of this emerging field will also be explored with the hope that such a combination will lead to a paradigm shift in cancer treatments.
Collapse
|
36
|
Zhang B, Shao CW, Zhou KM, Li Q, Duan YT, Yang YS, Zhu HL. A NIR-triggered multifunctional nanoplatform mediated by Hsp70 siRNA for chemo-hypothermal photothermal synergistic therapy. Biomater Sci 2021; 9:6501-6509. [PMID: 34582538 DOI: 10.1039/d1bm01006a] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recently, hypothermal photothermal therapy (HPTT) seemed essential for the future clinical transformation of cancer optical therapies. However, at a lower working temperature, heat shock proteins (HSPs) seriously affect the anti-tumor effect of HPTT. This work reports a reasonable design of a dual-responsive nanoplatform for the synergistic treatment of chemotherapy and HPTT. We adopted a one-step method to wrap indocyanine green (ICG) into imidazole skeleton-8 (ZIF-8) and further loaded it with the chemotherapy drug doxorubicin (DOX). Furthermore, we introduced Hsp-70 siRNA to block the affection of HSPs at an upstream node, thereby avoiding the side effects of traditional heat shock protein inhibitors. The prepared ZIF-8@ICG@DOX@siRNA nanoparticles (ZID-Si NPs) could significantly improve the stability of siRNA to effectively down-regulate the expression of HSP70 protein during the photothermal therapy, thus realizing the pH-controlled and NIR-triggered release of the chemotherapeutical drug DOX. Moreover, tumors were also imaged accurately by ICG wrapped in ZID-Si nanoparticles. After the evaluation of the in vitro and in vivo photothermal effect as well as the anti-tumor activity, we found that the added Hsp-70 siRNA enhanced the synergistic anti-cancer activity of HPTT and chemotherapy. In summary, this work holds great potential in cancer treatment, and suggests better efficacy of synergistic chemo/HPTT than the single-agent therapy.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.
| | - Chen-Wen Shao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.
| | - Kang-Min Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.
| | - Qin Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.
| | - Yong-Tao Duan
- Henan provincial key laboratory of children's genetics and metabolic diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, PR China
| | - Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.
| |
Collapse
|
37
|
Zhang W, An EK, Hwang J, Jin JO. Mice Plasmacytoid Dendritic Cells Were Activated by Lipopolysaccharides Through Toll-Like Receptor 4/Myeloid Differentiation Factor 2. Front Immunol 2021; 12:727161. [PMID: 34603298 PMCID: PMC8481683 DOI: 10.3389/fimmu.2021.727161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/02/2021] [Indexed: 11/29/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are known to respond to viral infections. However, the activation of pDCs by bacterial components such as lipopolysaccharides (LPS) has not been well studied. Here, we found that pDCs, conventional dendritic cells (cDCs), and B cells express high levels of toll-like receptor 4 (TLR4), a receptor for LPS. Moreover, LPS could effectively bind to not only cDCs but also pDCs and B cells. Intraperitoneal administration of LPS promoted activation of splenic pDCs and cDCs. LPS treatment led to upregulation of interferon regulatory factor 7 (IRF7) and induced production of interferon-alpha (IFN-α) in splenic pDCs. Furthermore, LPS-dependent upregulation of co-stimulatory molecules in pDCs did not require the assistance of other immune cells, such as cDCs. However, the production levels of IFN-α were decreased in cDC-depleted splenocytes, indicating that cDCs may contribute to the enhancement of IFN-α production in pDCs. Finally, we showed that activation of pDCs by LPS requires the TLR4 and myeloid differentiation factor 2 (MD2) signaling pathways. Thus, these results demonstrate that the gram-negative component LPS can directly stimulate pDCs via TLR4/MD2 stimulation in mice.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Eun-Koung An
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
38
|
Zhang W, An EK, Park HB, Hwang J, Dhananjay Y, Kim SJ, Eom HY, Oda T, Kwak M, Lee PCW, Jin JO. Ecklonia cava fucoidan has potential to stimulate natural killer cells in vivo. Int J Biol Macromol 2021; 185:111-121. [PMID: 34119543 DOI: 10.1016/j.ijbiomac.2021.06.045] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022]
Abstract
Fucoidan is a sulfated polysaccharide, derived from various marine brown seaweeds, that has immunomodulatory effects. In this study, we analyzed the effects of five different fucoidans, which were extracted from Ascophyllum nodosum, Undaria pinnatifida, Macrocystis pyrifera, Fucus vesiculosus, and Ecklonia cava, on natural killer (NK) cell activation in mice. Among these, E. cava fucoidan (ECF) promoted an increase in the number of NK cells in the spleen and had the strongest effect on the activation of NK cells. Additionally, we observed that DC stimulation was required for NK cell activation and that ECF had the most potent effect on splenic dendritic cells (DC). Finally, ECF treatment effectively prevented infiltration of CT-26 carcinoma cells in the lungs of BALB/c mice in an NK cell dependent manner. Collectively, these results suggest that ECF could be a suitable candidate for enhancing NK cell-mediated anti-cancer immunity.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Eun-Koung An
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea; Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hae-Bin Park
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea; Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea; Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Yadav Dhananjay
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - So-Jung Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hee-Yun Eom
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Tatsuya Oda
- Division of Biochemistry, Faculty of Fisheries, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, Nagasaki 852-8521, Japan
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - Peter Chang-Whan Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea.
| | - Jun-O Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea; Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
39
|
Zhang W, Hwang J, Yadav D, An EK, Kwak M, Lee PCW, Jin JO. Enhancement of Immune Checkpoint Inhibitor-Mediated Anti-Cancer Immunity by Intranasal Treatment of Ecklonia cava Fucoidan against Metastatic Lung Cancer. Int J Mol Sci 2021; 22:9125. [PMID: 34502035 PMCID: PMC8431244 DOI: 10.3390/ijms22179125] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/11/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in the lung was examined by intranasal administration. In C57BL/6 and BALB/c mice, intranasal administration of ECF promoted the activation of dendritic cells (DCs), natural killer (NK) cells, and T cells in the mediastinal lymph node (mLN). The ECF-induced NK and T cell activation was mediated by DCs. In addition, intranasal injection with ECF enhanced the anti-PD-L1 antibody-mediated anti-cancer activities against B16 melanoma and CT-26 carcinoma tumor growth in the lungs, which were required cytotoxic T lymphocytes and NK cells. Thus, these data demonstrated that ECF functioned as a mucosal adjuvant that enhanced the immunotherapeutic effect of immune checkpoint inhibitors against metastatic lung cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; (W.Z.); (J.H.)
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; (W.Z.); (J.H.)
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
| | - Eun-Koung An
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, Korea;
| | - Peter Chang-Whan Lee
- ASAN Medical Center, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China; (W.Z.); (J.H.)
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (D.Y.); (E.-K.A.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
40
|
Cheng HW, Ou YL, Kuo CC, Tsao HY, Lu HE. Ansamitocin P3-Loaded Gold-NanoCage Conjugated with Immune Checkpoint Inhibitor to Enhance Photo-Chemo-Thermal Maturation of Dendritic Cells for Hepatocellular Carcinoma. Polymers (Basel) 2021; 13:2726. [PMID: 34451265 PMCID: PMC8398096 DOI: 10.3390/polym13162726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022] Open
Abstract
Immunotherapy is a newly developed method for cancer treatment, but still generates limited response in partial patients for hepatocellular carcinoma (HCC) because the immunity cycle is limited by the tumor microenvironment (TME). Herein, we introduce multifunctional gold nanocages (AuNCs)-based nanocarriers with Ansamitocin P3 (AP3) loaded and anti-PDL1 binding (AP3-AuNCs-anti-PDL1) which can combine photothermal therapy, chemotherapeutic agent-triggered DCs maturation, and checkpoint immunotherapy in one platform. The AP3-AuNCs-anti-PDL1 using Avidin-biotin to bind anti-PDL1 on the surface of AP3-AuNCs showed specifically cellular targeting compared to AuNCs, which can increase the immune responses. The AP3-AuNCs+NIR-10 min exhibited the highly activated DCs maturation with two-fold higher than control+NIR, which can be attributed to the significant release of AP3. The results illustrated the synergistic effect of tumor-associated antigens (TAAs) and controlled AP3 release under near infrared (NIR) in triggering effective DCs maturation. Among them, AP3 release played the more important role than the TAAs under PTT in promoting T-cell activation. These results illustrate the promising potential of AuNCs-based nanocarriers combined with AP3 and the checkpoint inhibitors to strengthen the positive loop of immunity cycle.
Collapse
Affiliation(s)
- Hung-Wei Cheng
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan; (H.-W.C.); (Y.-L.O.); (C.-C.K.); (H.-Y.T.)
| | - Yu-Ling Ou
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan; (H.-W.C.); (Y.-L.O.); (C.-C.K.); (H.-Y.T.)
| | - Chia-Chi Kuo
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan; (H.-W.C.); (Y.-L.O.); (C.-C.K.); (H.-Y.T.)
| | - Hsin-Yi Tsao
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan; (H.-W.C.); (Y.-L.O.); (C.-C.K.); (H.-Y.T.)
| | - Huai-En Lu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu 300193, Taiwan
| |
Collapse
|
41
|
Near-infrared light-responsive liposomes for protein delivery: Towards bleeding-free photothermally-assisted thrombolysis. J Control Release 2021; 337:212-223. [PMID: 34284049 DOI: 10.1016/j.jconrel.2021.07.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022]
Abstract
Smart drug delivery systems represent state-of-the-art approaches for targeted therapy of life-threatening diseases such as cancer and cardiovascular diseases. Stimuli-responsive on-demand release of therapeutic agents at the diseased site can significantly limit serious adverse effects. In this study, we engineered a near-infrared (NIR) light-responsive liposomal gold nanorod-containing platform for on-demand delivery of proteins using a hybrid formulation of ultrasmall gold nanorods (AuNRs), thermosensitive phospholipid (DPPC) and non-ionic surfactant (Brij58). In light-triggered release optimization studies, 55.6% (± 4.8) of a FITC-labelled model protein, ovalbumin (MW 45 kDa) was released in 15 min upon NIR irradiation (785 nm, 1.35 W/cm2 for 5 min). This platform was then utilized to test on-demand delivery of urokinase-plasminogen activator (uPA) for bleeding-free photothermally-assisted thrombolysis, where the photothermal effect of AuNRs would synergize with the released uPA in clot lysis. Urokinase light-responsive liposomes showed 80.7% (± 4.5) lysis of an in vitro halo-clot model in 30 min following NIR irradiation (785 nm, 1.35 W/cm2 for 5 min) compared to 36.3% (± 4.4) and 15.5% (± 5.5) clot lysis from equivalent free uPA and non-irradiated liposomes respectively. These results show the potential of low-dose, site-specific thrombolysis via the combination of light-triggered delivery/release of uPA from liposomes combined with photothermal thrombolytic effects from gold nanorods. In conclusion, newly engineered, gold nanorod-based, NIR light-responsive liposomes represent a promising drug delivery system for site-directed, photothermally-stimulated therapeutic protein release.
Collapse
|
42
|
Hwang J, Zhang W, Park HB, Yadav D, Jeon YH, Jin JO. Escherichia coli adhesin protein-conjugated thermal responsive hybrid nanoparticles for photothermal and immunotherapy against cancer and its metastasis. J Immunother Cancer 2021; 9:jitc-2021-002666. [PMID: 34230112 PMCID: PMC8261870 DOI: 10.1136/jitc-2021-002666] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 12/21/2022] Open
Abstract
Background Advanced cancer therapy is targeted at primary tumors and also recurrent or metastatic cancers. Combinational cancer treatment has recently shown high efficiency against recurrent and metastatic cancers. In this study, we synthesized a thermal responsive hybrid nanoparticle (TRH) containing FimH, an immune stimulatory recombinant protein, for the induction of a combination of photothermal therapy (PTT) and immunotherapy against cancer and its metastasis. Methods The hybrid nanoparticle was incorporated with a near-infrared (NIR) absorbent, indocyanine green, and decorated with FimH on its surface to form F-TRH. F-TRH was evaluated for its anticancer and antimetastatic effects against CT-26 carcinoma in mice by combining PTT and immunotherapy. Results NIR laser irradiation elicited an elevation of temperature in F-TRH, which induced apoptosis in CT-26 carcinoma cells in vitro. In addition, F-TRH and NIR laser irradiation promoted photothermal-mediated therapeutic effects against CT-26 and 4T1 tumors in mice. The release of FimH from F-TRH in response to elevated temperature and apoptotic bodies of cancer cells via PTT elicited dendritic cell-mediated cancer antigen-specific T-cell responses, which subsequently inhibited the second challenge of CT-26 and 4T1 cell growth in the lung. Conclusions These data demonstrate the potential use of F-TRH for immuno-photothermal therapy against cancer and its recurrence and metastasis.
Collapse
Affiliation(s)
- Juyoung Hwang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hae-Bin Park
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China .,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
43
|
Huang X, Lu Y, Guo M, Du S, Han N. Recent strategies for nano-based PTT combined with immunotherapy: from a biomaterial point of view. Theranostics 2021; 11:7546-7569. [PMID: 34158866 PMCID: PMC8210617 DOI: 10.7150/thno.56482] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/23/2021] [Indexed: 01/04/2023] Open
Abstract
Cancer has been a great threat to humans for decades. Due to the limitations of monotherapy, combinational therapies such as photothermal therapy (PTT) and immunotherapy have gained increasing attention with expectation to overcome the shortfalls of each other and obtain satisfactory therapeutic outcomes. PTT can inhibit primary tumors by thermal ablation but usually fails to achieve complete eradication and cannot prevent metastasis and recurrence. Meanwhile, the efficacy of immunotherapy is usually attenuated by the weak immunogenicity of tumor and the immunosuppressive tumor microenvironment (ITM). Therefore, many recent studies have attempted to synergize PTT with immunotherapy in order to enhance the therapeutic efficacy. In this review, we aim to summarize the cutting-edge strategies in combining nano-based PTT with immunotherapy for cancer treatment. Herein, the combination strategies were mainly classified into four categories, including 1) nano-based PTT combined with antigens to induce host immune responses; 2) nano-based PTT in combination with immune adjuvants acting as in situ vaccines; 3) nano-based PTT synergized with immune checkpoint blockade or other regulators to relieve the ITM; 4) nano-based PTT combined with CAR-T therapy or cytokine therapy for tumor treatment. The characteristics of various photothermal agents and nanoplatforms as well as the immunological mechanisms for the synergism were also introduced in detail. Finally, we discussed the existing challenges and future prospects in combined PTT and immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Shouying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ning Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
44
|
Yu N, Ding M, Li J. Near-Infrared Photoactivatable Immunomodulatory Nanoparticles for Combinational Immunotherapy of Cancer. Front Chem 2021; 9:701427. [PMID: 34109160 PMCID: PMC8181730 DOI: 10.3389/fchem.2021.701427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 12/17/2022] Open
Abstract
As a promising treatment option for cancer, immunotherapy can eliminate local and distant metastatic tumors and even prevent recurrence through boosting the body’s immune system. However, immunotherapy often encounters the issues of limited therapeutic efficacy and severe immune-related adverse events in clinical practices, which should be mainly due to the non-specific accumulations of immunotherapeutic agents. Activatable immunomodulatory agents that are responsive to endogenous stimuli in tumor microenvironment can afford controlled immunotherapeutic actions, while they still face certain extent of off-target activation. Since light has the advantages of noninvasiveness, simple controllability and high spatio-temporal selectivity, therapeutic agents that can be activated by light, particularly near-infrared (NIR) light with minimal phototoxicity and strong tissue penetrating ability have been programmed for cancer treatment. In this mini review, we summarize the recent progress of NIR photoactivatable immunomodulatory nanoparticles for combinational cancer immunotherapy. The rational designs, constructions and working mechanisms of NIR photoactivatable agents are first briefly introduced. The uses of immunomodulatory nanoparticles with controlled immunotherapeutic actions upon NIR photoactivation for photothermal and photodynamic combinational immunotherapy of cancer are then summarized. A conclusion and discussion of the existing challenges and further perspectives for the development and clinical translation of NIR photoactivatable immunomodulatory nanoparticles are finally given.
Collapse
Affiliation(s)
- Ningyue Yu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Mengbin Ding
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Jingchao Li
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| |
Collapse
|
45
|
Yu H, Ji M. Recent Advances of Organic Near-Infrared II Fluorophores in Optical Properties and Imaging Functions. Mol Imaging Biol 2021; 23:160-172. [PMID: 33030708 DOI: 10.1007/s11307-020-01545-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 12/30/2022]
Abstract
Near-infrared (NIR) fluorescence imaging (FI) has become a research hotspot because of its distinctive imaging properties: high temporal resolution and sensitivity. Especially in recent years, with the research focus of NIR FI shifting to the NIR-II region, which has better imaging performance, it is expected that NIR FI will find significant applications in the field of in vivo imaging. One of the most crucial directions for research into NIR-II FI is the promotion of novel NIR-II fluorophores with superior imaging properties. The remarkable advantages of organic NIR-II fluorophores in biosafety make them more promising than other fluorescent materials in certain applications. But serious defects in their fluorescence performance preclude particular imaging effects and limit imaging functions. In this review, we summarize and discuss the recent leading literature on overcoming the defects of organic NIR-II fluorophores, demonstrating the potential for further improving their imaging properties. In addition, we cover the functions of NIR-II FI that are promoted by the development of fluorophores, notably including its outlook on molecular imaging in vivo.
Collapse
Affiliation(s)
- Haoli Yu
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Min Ji
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
46
|
Advances in Nanomaterial-Mediated Photothermal Cancer Therapies: Toward Clinical Applications. Biomedicines 2021; 9:biomedicines9030305. [PMID: 33809691 PMCID: PMC8002224 DOI: 10.3390/biomedicines9030305] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/14/2021] [Indexed: 12/24/2022] Open
Abstract
Photothermal therapy (PTT) has attracted extensive research attention as a noninvasive and selective treatment strategy for numerous cancers. PTT functions via photothermal effects induced by converting light energy into heat on near-infrared laser irradiation. Despite the great advances in PTT for cancer treatment, the photothermal therapeutics using laser devise only or non-specific small molecule PTT agents has been limited because of its low photothermal conversion efficiency, concerns about the biosafety of the photothermal agents, their low tumor accumulation, and a heat resistance of specific types of cancer. Using nanomaterials as PTT agents themselves, or for delivery of PTT agents, offers improved therapeutic outcomes with fewer side effects through enhanced photothermal conversion efficiency, accumulation of the PTT agent in the tumor tissue, and, by extension, through combination with other therapies. Herein, we review PTT’s current clinical progress and present the future outlooks for clinical applications. To better understand clinical PTT applications, we describe nanomaterial-mediated photothermal effects and their mechanism of action in the tumor microenvironment. This review also summarizes recent studies of PTT alone or in combination with other therapies. Overall, innovative and strategically designed PTT platforms are promising next-generation noninvasive cancer treatments to move closer toward clinical applications.
Collapse
|
47
|
Xu PY, Zheng X, Kankala RK, Wang SB, Chen AZ. Advances in Indocyanine Green-Based Codelivery Nanoplatforms for Combinatorial Therapy. ACS Biomater Sci Eng 2021; 7:939-962. [PMID: 33539071 DOI: 10.1021/acsbiomaterials.0c01644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Indocyanine green (ICG), a near-infrared (NIR) agent with an excellent imaging performance, has captivated enormous interest from researchers owing to its excellent therapeutic and imaging abilities. Although various nanoplatforms-based drug delivery systems (DDS) with the ability to overcome the clinical limitations of ICG has been reported, ICG-medicated conventional cancer diagnosis and photorelated therapies still lack in exhibiting the therapeutic efficacy, resulting in incomplete or partly tumor elimination. In the view of addressing these concerns, various DDSs have been engineered for the efficient codelivery of combined therapeutic agents with ICG, aiming to achieve promising therapeutic results due to multifunctional imaging-guided synergistic antitumor effects. In this article, we will systematically review currently available nanoplatforms based on polymers, inorganic, proteins, and metal-organic frameworks (MOFs), among others, for codelivery of ICG along with other therapeutic agents, providing a foundation for future clinical development of ICG. In addition, codelivery systems for ICG and different mechanism-based therapeutic agents will be illustrated. In summary, we conclude the review with the challenges and perspectives of ICG-based versatile nanoplatforms in detail.
Collapse
Affiliation(s)
- Pei-Yao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Xiang Zheng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| |
Collapse
|
48
|
Brouillard A, Deshpande N, Kulkarni AA. Engineered Multifunctional Nano- and Biological Materials for Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2001680. [PMID: 33448159 DOI: 10.1002/adhm.202001680] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy is set to emerge as the future of cancer therapy. However, recent immunotherapy trials in different cancers have yielded sub-optimal results, with durable responses seen in only a small fraction of patients. Engineered multifunctional nanomaterials and biological materials are versatile platforms that can elicit strong immune responses and improve anti-cancer efficacy when applied to cancer immunotherapy. While there are traditional systems such as polymer- and lipid-based nanoparticles, there is a wide variety of other materials with inherent and additive properties that can allow for more potent activation of the immune system. By synthesizing and applying multifunctional strategies, it allows for a more extensive and more effective repertoire of tools to use in the wide variety of situations that cancer presents itself. Here, several types of nanoscale and biological material strategies and platforms that provide their inherent benefits for targeting and activating multiple aspects of the immune system are discussed. Overall, this review aims to provide a comprehensive understanding of recent advances in the field of multifunctional cancer immunotherapy and trends that pave the way for more diverse and tactical regression of tumors through soliciting responses by either the adaptive or innate immune system, and even both simultaneously.
Collapse
Affiliation(s)
- Anthony Brouillard
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Nilesh Deshpande
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Ashish A. Kulkarni
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
49
|
Yu Y, Cheng Y, Tong J, Zhang L, Wei Y, Tian M. Recent advances in thermo-sensitive hydrogels for drug delivery. J Mater Chem B 2021; 9:2979-2992. [DOI: 10.1039/d0tb02877k] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thermo-sensitive hydrogels based on different polymers have been broadly used in the pharmaceutical fields. In this review, the state-of-the-art thermo-sensitive hydrogels for drug delivery are elaborated
Collapse
Affiliation(s)
- Yibin Yu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences
- Changchun 130022
- China
| | - Yi Cheng
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences
- Changchun 130022
- China
| | - Junye Tong
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences
- Changchun 130022
- China
| | - Lei Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences
- Changchun 130022
- China
| | - Yen Wei
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University
- Beijing 100084
- China
| | - Mei Tian
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou
- Zhejiang, 310009
- China
| |
Collapse
|
50
|
Polysaccharide from Codium fragile Induces Anti-Cancer Immunity by Activating Natural Killer Cells. Mar Drugs 2020; 18:md18120626. [PMID: 33302530 PMCID: PMC7763488 DOI: 10.3390/md18120626] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/21/2022] Open
Abstract
Natural polysaccharides exhibit beneficial immune modulatory effects, including immune stimulatory and anti-cancer activities. In this study, we examined the effect of Codium fragile polysaccharide (CFP) on natural killer (NK) cell activation, and its effect on tumor-bearing mice. Intravenous CFP treatment of C57BL/6 mice resulted in the upregulation of CD69, which is a marker associated with NK cell activation. In addition, intracellular levels of interferon (IFN)-γ and the cytotoxic mediators perforin and granzyme B were markedly increased in response to the CFP treatment of splenic NK cells. IFN-γ production by NK cells was directly induced by CFP, whereas the upregulation of CD69 and cytotoxic mediators required IL-12. Finally, intraperitoneal treatment with CFP prevented CT-26 (murine carcinoma) tumor cell infiltration in the lungs, without significantly reducing the body weight. In addition, treatment with CFP prevented B16 melanoma cell infiltration in the lung of C57BL/6 mice. Moreover, the anti-tumor effect was diminished by the depletion of NK cells. Therefore, these data suggest that CFP may be used as an NK cell stimulator to produce a phenomenon that contributes to anti-cancer immunity.
Collapse
|