1
|
Lu Y, Xie X, Luo L. Ferroptosis crosstalk in anti-tumor immunotherapy: molecular mechanisms, tumor microenvironment, application prospects. Apoptosis 2024; 29:1914-1943. [PMID: 39008197 DOI: 10.1007/s10495-024-01997-8] [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] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Immunotherapies for cancer, specifically immune checkpoint inhibition (ICI), have shown potential in reactivating the body's immune response against tumors. However, there are challenges to overcome in addressing drug resistance and improving the effectiveness of these treatments. Recent research has highlighted the relationship between ferroptosis and the immune system within immune cells and the tumor microenvironment (TME), suggesting that combining targeted ferroptosis with immunotherapy could enhance anti-tumor effects. This review explores the potential of using immunotherapy to target ferroptosis either alone or in conjunction with other therapies like immune checkpoint blockade (ICB) therapy, radiotherapy, and nanomedicine synergistic treatments. It also delves into the roles of different immune cell types in promoting anti-tumor immune responses through ferroptosis. Together, these findings provide a comprehensive understanding of synergistic immunotherapy focused on ferroptosis and offer innovative strategies for cancer treatment.
Collapse
Affiliation(s)
- Yining Lu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xiaoting Xie
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
2
|
Iaconisi GN, Ahmed A, Lauria G, Gallo N, Fiermonte G, Cowman MK, Capobianco L, Dolce V. Targeting mitochondria in Cancer therapy: Machine learning analysis of hyaluronic acid-based drug delivery systems. Int J Biol Macromol 2024; 283:137840. [PMID: 39566768 DOI: 10.1016/j.ijbiomac.2024.137840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Mitochondrial alterations play a crucial role in the development and progression of cancer. Dysfunctional mitochondria contribute to the acquisition of key hallmarks of cancer, including sustained proliferative signaling, evasion of growth suppressors, and resistance to cell death. Consequently, targeting mitochondrial dysfunction has emerged as a promising therapeutic strategy. Hyaluronic acid (HA), a naturally occurring glycosaminoglycan, has garnered significant attention due to its multifaceted roles in cancer biology. METHODS We employed a Systematic Literature Review (SLR) approach to examine a collection of 90 scientific publications using a text mining technique leveraging the Latent Dirichlet Allocation (LDA) algorithm. RESULTS The result of this activity, performed through the MySLR digital platform, allowed us to identify a set of two distinct topics representing the research domain. Specifically, Topic 1 comprised 41 papers, while Topic 2 comprised 49 papers. CONCLUSIONS The computational analysis highlighted that the integration of HA into drug delivery systems represents a promising approach to enhance the effectiveness and safety of cancer therapies. The discussed clinical trials provided compelling evidence of the potential of HA-based treatments in targeting cancer cells while minimizing adverse effects on healthy tissues.
Collapse
Affiliation(s)
- Giorgia Natalia Iaconisi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy.
| | - Graziantonio Lauria
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Cosenza, Italy.
| | - Nunzia Gallo
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Giuseppe Fiermonte
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy.
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, NY, New York, USA; Department of Orthopedic Surgery, Grossman School of Medicine, New York University, NY, New York, USA.
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Vincenza Dolce
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Cosenza, Italy.
| |
Collapse
|
3
|
Al-Taie A, Özcan Bülbül E. A paradigm use of monoclonal antibodies-conjugated nanoparticles in breast cancer treatment: current status and potential approaches. J Drug Target 2024; 32:45-56. [PMID: 38096045 DOI: 10.1080/1061186x.2023.2295803] [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: 08/23/2023] [Accepted: 12/12/2023] [Indexed: 12/30/2023]
Abstract
Monoclonal antibodies (mAbs) are integral to cancer treatment over conventional non-specific therapy methods. This study provides a scoping review of the clinically approved mAbs, focusing on the current application of different nanocarrier technologies as drug delivery targets for mAb-conjugated nanoparticles (NPs) as potential features for breast cancer (BC) treatment. An extensive literature search was conducted between the years 2000 and 2023 using various sources of databases. The first part covered mAb classification, types, and mechanisms of action, pharmacokinetics and clinical applications in BC. The second part covered polymeric, lipid and inorganic-based NPs, which are a variety of mAb-conjugated NPs targeting BC. A total of 20 relevant studies were enrolled indicating there are three different types of nanoparticular systems (polymeric NPs, inorganic NPs and lipid-based NPs) that can be used for BC treatment by being loaded with various active substances and conjugated with these antibodies. While mAbs have altered the way in cancer treatment due to targeting cancer cells specifically, the delivery of mAbs with nanoparticulate systems is important in the treatment of BC, as NPs are still being investigated as distinctive and promising drug delivery methods that can be employed for effective treatment of BC.
Collapse
Affiliation(s)
- Anmar Al-Taie
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| | - Ece Özcan Bülbül
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istinye University, Istanbul, Türkiye
| |
Collapse
|
4
|
Babaei Khorzoughi S, Tavakoli M, Mortazavi M, Jafarnejad Z, Malekpour A, Kopaiee Malek T, Kargar F. A review of recombinant HER3 affibodies with an effective diagnostic view of cancer cells. J Drug Target 2024:1-12. [PMID: 39485069 DOI: 10.1080/1061186x.2024.2420202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024]
Abstract
Breast cancer is one of the leading causes of cancer-related deaths among women globally. Factors like increased expression of HER family members contribute to its development, with elevated HER3 levels-especially in conjunction with tyrosine kinase receptors like HER2-playing a critical role in activating cancer pathways essential for cell survival and proliferation. Detecting high HER3 levels is vital for effective treatment. Affibody proteins, a class that includes antibodies, are used to identify elevated HER3 expression due to their high binding affinity. These innovative non-immune probes show promise in therapy, diagnostics, and biotechnology because of their exceptional specificity and affinity for target proteins. The design of recombinant affibodies enhances HER3 detection accuracy and supports the development of targeted therapies. Advanced engineering techniques optimize these affibodies for stability and binding efficacy, making them suitable for clinical applications. Additionally, their versatility allows integration with imaging technologies for real-time monitoring of HER3 expression and therapeutic responses. This comprehensive approach could lead to more personalized treatment options for patients with HER3-positive breast cancers, improving patient management and outcomes. This study presents recombinant affibodies designed to bind HER3 for cancer cell identification and introduces novel methods for producing various affibody molecules.
Collapse
Affiliation(s)
- Sahar Babaei Khorzoughi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Mehrnoosh Tavakoli
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Mojtaba Mortazavi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Zahra Jafarnejad
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | | | - Tara Kopaiee Malek
- Department of Cell and Molecular Biology, Faculty of Science, Azad University of Damghan, Damghan, Iran
| | - Farzane Kargar
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Pinho S, Coelho JMP, Gaspar MM, Reis CP. Advances in localized prostate cancer: A special focus on photothermal therapy. Eur J Pharmacol 2024; 983:176982. [PMID: 39260812 DOI: 10.1016/j.ejphar.2024.176982] [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: 04/18/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
Prostate cancer (PCa) is a high prevalence disease, per 10000 habitants, that tends to increase with age. This pathology is difficult to detect at an early stage due to the absence of symptoms, hence the importance of monitoring signs for early detection. This disease can be detected by various methods, including plasmatic levels of prostate-specific antigen (PSA) and rectal touch, with biopsy being necessary to confirm the diagnosis. Patients affected by prostate cancer can have localized or advanced disease. There are conventional approaches that have been used as a reference in localized cancer, such as active surveillance, surgery, or radiotherapy. However, the adverse effects might vary and, sometimes, they can be permanent. An overview about the innovative therapeutic approaches to improve outcomes in terms of both tumor remission and side effects for localized PCa is presented. In case of emerging light-based treatment strategies, they aimed at ablating tumor tissue by inducing an external light are non-invasive, localized and, considerably, they are able to reduce lesions in peripheral tissues. One is photodynamic therapy (PDT) and it involves the photooxidation of molecules culminating in the formation of reactive oxygen species (ROS), inducing cell death. On the other hand, photothermal therapy (PTT) is based on inducing hyperthermia in cancer cells by irradiating them with beams of light at a specific wavelength. To improve the heat generated, gold nanoparticles (AuNPs) have those desirable characteristics that have drawn attention to PTT. Various studies point to AuNPs as efficient nanomaterials in PTT for the treatment of tumors, including prostate cancer. This review includes the most representative advances in this research field, dated from 1998 to 2023. It is noticed that several advances have been made and the way to find the effective treatment without impacting adverse side effects is shorter.
Collapse
Affiliation(s)
- Sara Pinho
- Research Institute for Medicines, IMed.ULisboa - Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - João M P Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, 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; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, 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
|
6
|
Biray Avci C, Goker Bagca B, Nikanfar M, Takanlou LS, Takanlou MS, Nourazarian A. Tumor microenvironment and cancer metastasis: molecular mechanisms and therapeutic implications. Front Pharmacol 2024; 15:1442888. [PMID: 39600368 PMCID: PMC11588459 DOI: 10.3389/fphar.2024.1442888] [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: 06/03/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and metastasis. This review summarizes the current research on how the TME promotes metastasis through molecular pathways, focusing on key components, such as cancer-associated fibroblasts, immune cells, endothelial cells, cytokines, and the extracellular matrix. Significant findings have highlighted that alterations in cellular communication within the TME enable tumor cells to evade immune surveillance, survive, and invade other tissues. This review highlights the roles of TGF-β and VEGF signaling in promoting angiogenesis and extracellular matrix remodeling, which facilitate metastasis. Additionally, we explored how metabolic reprogramming of tumor and stromal cells, influenced by nutrient availability in the TME, drives cancer progression. This study also evaluated the therapeutic strategies targeting these interactions to disrupt metastasis. By providing a multidisciplinary perspective, this study suggests that understanding the molecular basis of the TME can lead to more effective cancer therapies and identify potential avenues for future research. Future research on the TME should prioritize unraveling the molecular and cellular interactions within this complex environment, which could lead to novel therapeutic strategies and personalized cancer treatments. Moreover, advancements in technologies such as single-cell analysis, spatial transcriptomics, and epigenetic profiling offer promising avenues for identifying new therapeutic targets and improving the efficacy of immunotherapies, particularly in the context of metastasis.
Collapse
Affiliation(s)
- Cigir Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Türkiye
| | - Bakiye Goker Bagca
- Department of Medical Biology, Faculty of Medicine, Adnan Menderes University, Aydin, Türkiye
| | - Masoud Nikanfar
- Department of Neurology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
- Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
7
|
Liu K, Hoover AR, Wang L, Sun Y, Valerio TI, Furrer C, Adams J, Yang J, Li M, Chen WR. Localized ablative immunotherapy enhances antitumor immunity by modulating the transcriptome of tumor-infiltrating Gamma delta T cells. Cancer Lett 2024; 604:217267. [PMID: 39307410 PMCID: PMC11471373 DOI: 10.1016/j.canlet.2024.217267] [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: 06/28/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Gamma delta T cells (γδT cells) play crucial roles in the immune response against tumors, yet their functional dynamics under different cancer therapies remain poorly understood. Laser Ablative Immunotherapy (LAIT) is a novel cancer treatment modality combining local photothermal therapy (PTT) and intratumoral injection of an immunostimulant, N-dihydrogalactochitosan (glycated chitosan, GC). LAIT has been shown to induce systemic antitumor immune responses in pre-clinical studies and clinical trials, eradicating both treated local tumors and untreated distant metastases. In this study, we used LAIT to treat breast tumors in a mouse model and investigated the effects of LAIT on tumor-infiltrating γδT cells using single-cell RNA sequencing (scRNAseq). We characterized the γδT cells from tumors in control, PTT, GC, and LAIT (PTT + GC) groups, by identifying six distinct subtypes: activated, cytotoxic, cycling cytotoxic, IFN-enriched, antigen-presenting, and IL17-producing γδT cells. Differential gene expression analysis revealed that LAIT significantly upregulated genes associated with T cell activation, leukocyte adhesion, and interferon signaling in treated tumor tissues while downregulating genes involved in protein folding and stress responses. LAIT also uniquely increased the proportion of IL17-producing γδT cells, which correlated with prolonged survival in breast cancer patients, as analyzed using TCGA data. Furthermore, the transcriptomic profiles of γδT cells in LAIT-treated tumors closely resembled those in immune checkpoint inhibitor (ICI)-treated patients, suggesting potential synergistic effects. Our findings indicate that LAIT modulates the γδT cell transcriptome, enhancing their antitumor capabilities and providing a basis for combining LAIT with ICI therapy to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
8
|
Alsaidan OA, Elkomy MH, Zaki RM, Tulbah AS, Yusif RM, Eid HM. Brain targeting of venlafaxine via intranasal transbilosomes thermogel for improved management of depressive disorder. J Pharm Sci 2024; 113:3304-3314. [PMID: 39216538 DOI: 10.1016/j.xphs.2024.08.026] [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: 06/22/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The current research aimed to design and optimize hyaluronic acid-coated transbilosomes containing venlafaxine (VLF-HA-TBLs) for nose-to-brain delivery for improved management of depressive disorder. Venlafaxine-loaded transbilosomes (VLF-TBLs) were developed according to the film hydration procedure, optimized for maximum efficiency using the quality by design-based Box-Behnken design (BBD), and then coated with hyaluronic acid (HA). The optimized VLF-HA-TBLs were subjected to in vitro characterization, integrated into a thermolabile gel, and then exposed to in vivo evaluation studies. The results revealed that the VLF-HA-TBLs formulation exhibited acceptable size (185.6 ± 4.9 nm), surface charge (-39.8 ± 1.7 mV), and entrapment efficiency (69.6 ± 2.6 %). The morphological study revealed that nanovesicles were spherical and displayed a consistent size distribution without particle aggregation. It also showed improved ex vivo nasal diffusion and a prolonged release profile. In addition, the formulated VLF-HA-TBLs were stable under the studied conditions and tolerable when applied intranasally. Compared to the intranasal administration of VLF solution (VLF-SOL), the biodistribution analysis showed that VLF-HA-TBLs delivered intranasally had a relative bioavailability of 441 % in the brain and 288 % in plasma. Moreover, the intranasal delivery of VLF-HA-TBLs demonstrated much higher bioavailability (512 %) in the brain compared to VLF-SOL administered intravenously. Collectively, it could be possible to infer that HA-TBLs might be an effective nanocarrier to administer VLF to the brain via the nasal route.
Collapse
Affiliation(s)
- Omar A Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, 72341, Saudi Arabia.
| | - Mohammed H Elkomy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, 72341, Saudi Arabia.
| | - Randa Mohammed Zaki
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62511, Egypt.
| | - Alaa S Tulbah
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm al Qura University, Makkah 21955, Saudi Arabia.
| | - Rehab Mohammad Yusif
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmaceutics and Pharmaceutical Industries, College of Pharmacy, Taibah University, P.O. Box 30039, Al-Madinah Al-Munawarah 41477, Saudi Arabia.
| | - Hussein M Eid
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62511, Egypt.
| |
Collapse
|
9
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
10
|
Marjani AA, Nader ND, Aghanejad A. Exosomes as targeted diagnostic biomarkers: Recent studies and trends. Life Sci 2024; 354:122985. [PMID: 39151882 DOI: 10.1016/j.lfs.2024.122985] [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: 04/12/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Different categories of extracellular vesicles (EVs) are identified based on their origin and formation processes. Among these, exosomes (EXOs) originate from endosomal compartments merging with the plasma membrane, forming small lipid vesicles that transport a range of molecular cargo such as nucleic acids, proteins, and lipids. The composition of EXOs varies depending on their cellular source, encompassing various cell types, including neutrophils, dendritic cells, and even tumor cells. Remarkably, EXOs possess inherent stability, low immunogenicity, and compatibility, making them efficient nano vectors for drug delivery. Imaging techniques like bioluminescence, fluorescence, and nuclear imaging are crucial in non-invasively tracking EXOs within living organisms. This process requires the attachment of radionuclides to the EXO's structure without altering its essential characteristics. Real-time imaging of EXOs is vital for their clinical application, and recent advancements in labeling and tracking methodologies provide insights into biodistribution, functionality, and potential pathways for EXO-mediated drug delivery. This review presents updated progress in the diverse applications of EXOs in targeted imaging across various modalities, where they function as contrast agents facilitating tissue visualization and disease tracking. Consequently, EXOs emerge as promising entities in medical diagnostics and imaging.
Collapse
Affiliation(s)
- Aida Abbasi Marjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, United States of America
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Guo M, Sun Y, Wang X, Wang Z, Yuan X, Chen X, Yuan X, Wang L. The MCIB Model: A Novel Theory for Describing the Spatial Heterogeneity of the Tumor Microenvironment. Int J Mol Sci 2024; 25:10486. [PMID: 39408814 PMCID: PMC11476373 DOI: 10.3390/ijms251910486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The tumor microenvironment (TME) can be regarded as a complex and dynamic microecosystem generated by the interactions of tumor cells, interstitial cells, the extracellular matrix, and their products and plays an important role in the occurrence, progression and metastasis of tumors. In a previous study, we constructed an IEO model (prI-, prE-, and pOst-metastatic niche) according to the chronological sequence of TME development. In this paper, to fill the theoretical gap in spatial heterogeneity in the TME, we defined an MCIB model (Metabolic, Circulatory, Immune, and microBial microenvironment). The MCIB model divides the TME into four subtypes that interact with each other in terms of mechanism, corresponding to the four major links of metabolic reprogramming, vascular remodeling, immune response, and microbial action, providing a new way to assess the TME. The combination of the MCIB model and IEO model comprehensively depicts the spatiotemporal evolution of the TME and can provide a theoretical basis for the combination of clinical targeted therapy, immunotherapy, and other comprehensive treatment modalities for tumors according to the combination and crosstalk of different subtypes in the MCIB model and provide a powerful research paradigm for tumor drug-resistance mechanisms and tumor biological behavior.
Collapse
Affiliation(s)
- Minghao Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Yinan Sun
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.S.)
| | - Xiaohui Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Zikun Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.S.)
| | - Xun Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| |
Collapse
|
12
|
Réthi-Nagy Z, Juhász S. Microbiome's Universe: Impact on health, disease and cancer treatment. J Biotechnol 2024; 392:161-179. [PMID: 39009231 DOI: 10.1016/j.jbiotec.2024.07.002] [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: 04/23/2024] [Revised: 05/27/2024] [Accepted: 07/07/2024] [Indexed: 07/17/2024]
Abstract
The human microbiome is a diverse ecosystem of microorganisms that reside in the body and influence various aspects of health and well-being. Recent advances in sequencing technology have brought to light microbial communities in organs and tissues that were previously considered sterile. The gut microbiota plays an important role in host physiology, including metabolic functions and immune modulation. Disruptions in the balance of the microbiome, known as dysbiosis, have been linked to diseases such as cancer, inflammatory bowel disease and metabolic disorders. In addition, the administration of antibiotics can lead to dysbiosis by disrupting the structure and function of the gut microbial community. Targeting strategies are the key to rebalancing the microbiome and fighting disease, including cancer, through interventions such as probiotics, fecal microbiota transplantation (FMT), and bacteria-based therapies. Future research must focus on understanding the complex interactions between diet, the microbiome and cancer in order to optimize personalized interventions. Multidisciplinary collaborations are essential if we are going to translate microbiome research into clinical practice. This will revolutionize approaches to cancer prevention and treatment.
Collapse
Affiliation(s)
- Zsuzsánna Réthi-Nagy
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged H-6728, Hungary
| | - Szilvia Juhász
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged H-6728, Hungary.
| |
Collapse
|
13
|
Zhou D, Li M, Wu W, Wu Y, Nong Q, Wang S, Hong R. Distribution characteristics of immune infiltration and lymphovascular invasion in patients with breast cancer skin recurrence. Cancer Immunol Immunother 2024; 73:223. [PMID: 39235656 PMCID: PMC11377393 DOI: 10.1007/s00262-024-03783-6] [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: 06/04/2024] [Accepted: 07/17/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND To assess the distribution characteristics of immune infiltration and lymphovascular invasion in breast cancer skin recurrence patients. METHODS We retrospectively analyzed the clinicopathological data of patients who underwent radical surgery for primary breast cancer and experienced skin recurrence between January 2001 and April 2019. Immune and lymphovascular biomarkers were quantified in primary breast cancers, skin lesions and visceral metastatic lesions. Differences in biomarkers distribution between matched tissues were statistically analyzed using the Wilcoxon signed-rank test and Kruskal-Wallis one-way ANOVA. RESULTS A total of 71 female breast cancer patients were reviewed in this study. Our study found that the expression levels of various lymphocyte immune markers in primary tumor specimens were higher than those in skin recurrences. The expression of CD8, CD57 and CD31 in primary breast cancer was higher than those in the skin. Compared to visceral metastatic lesions, D2-40 was highly expressed in the skin, while CD8 tended to decrease. In the skin specimens, the expression of CD8 (P < 0.001), FOXP3 (P = 0.006) and CD68 (P < 0.001) in the intratumoral area was higher, while the expression of CD57 (P < 0.001) was higher in the peritumoral area. Analyzing specimens from the same patient at different time points of skin progression, it was found that the expression of peritumoral CD4 decreased (P = 0.044) as the disease progressed. The low expression of D2-40 and CD163 in the skin lesions suggested a decrease in DFS. CONCLUSION The immune microenvironment of breast cancer skin recurrence may be in a state of suppression, and this suppression may intensify with disease progression. The pattern of skin recurrence may be more inclined toward lymphatic invasion. Our study provides new insights into the biological behaviors of this disease and its response to immunotherapy.
Collapse
Affiliation(s)
- Danyang Zhou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Mei Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China
| | - Wei Wu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China
| | - Ying Wu
- Department of Interventional Therapy, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qiaohong Nong
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Shusen Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China.
| | - Ruoxi Hong
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China.
| |
Collapse
|
14
|
Liu Y, Ding L, Chen G, Wang P, Wang X. A thermo-sensitive hydrogel with prominent hemostatic effect prevents tumor recurrence via anti-anoikis-resistance. J Nanobiotechnology 2024; 22:496. [PMID: 39164723 PMCID: PMC11334358 DOI: 10.1186/s12951-024-02739-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Tumor cells can survive when detached from the extracellular matrix (ECM) or lose cell-cell connections, a phenomenon known as anoikis-resistance (AR). AR is closely associated with tumor cell metastasis and recurrence, enabling tumor cells to disseminate, migrate, and invade after detachment. To address this issue, a novel intervention method combining intraoperative hemostasis with multifunctional nanozyme driven-enhanced chemodynamic therapy (ECDT) has been proposed, which holds the potential to weaken the AR capability of tumor cells and suppress tumor recurrence. Here, a nanocomposite containing a dendritic mesoporous nanoframework with Cu2+ was developed using an anion-assisted approach after surface PEG grafting and glucose oxidase (GOx) anchoring (DMSN-Cu@GOx/PEG). DMSN-Cu@GOx/PEG was further encapsulated in a thermal-sensitive hydrogel (H@DMSN-Cu@GOx/PEG). DMSN-Cu@GOx/PEG utilizes its high peroxidase (POD) activity to elevate intracellular ROS levels, thereby weakening the AR capability of bladder cancer cells. Additionally, through its excellent catalase (CAT) activity, DMSN-Cu@GOx/PEG converts the high level of hydrogen peroxide (H2O2) catalyzed by intracellular GOx into oxygen (O2), effectively alleviating tumor hypoxia, downregulating hypoxia-inducible factor-1α (HIF-1α) expression, inhibiting epithelial-mesenchymal transition (EMT) processes, and ultimately suppressing the migration and invasion of bladder cancer cells. Interestingly, in vivo results showed that the thermosensitive hydrogel H@DMSN-Cu@GOx/PEG could rapidly gel at body temperature, forming a gel film on wounds to eliminate residual tumor tissue after tumor resection surgery. Importantly, H@DMSN-Cu@GOx/PEG exhibited excellent hemostatic capabilities, effectively enhancing tissue coagulation during post-tumor resection surgery and mitigating the risk of cancer cell dissemination and recurrence due to surgical bleeding. Such hydrogels undoubtedly possess strong surgical application. Our developed novel nanosystem and hydrogel can inhibit the AR capability of tumor cells and prevent recurrence post-surgery. This study represents the first report of using dendritic mesoporous silica-based nanoreactors for inhibiting the AR capability of bladder cancer cells and suppressing tumor recurrence post-surgery, providing a new avenue for developing strategies to impede tumor recurrence after surgery.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Lei Ding
- School of Rare Earths, University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, People's Republic of China
| | - Gaojie Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Peiyuan Wang
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research On the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, People's Republic of China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China.
- Medical Research Institute, Wuhan University, Wuhan, 430071, People's Republic of China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
15
|
Guigue PA, Brezinov Y, Yasmeen A, Mbarik M, Salvador S, Lau S, Gotlieb WH, Brodeur MN. Ascites and Serum Interleukin-10 Levels as a Prognostic Tool for Ovarian Cancer Outcomes. Cancers (Basel) 2024; 16:2840. [PMID: 39199610 PMCID: PMC11352926 DOI: 10.3390/cancers16162840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Interleukin-10 (IL-10) has been shown to be present at high levels in the ascites of ovarian cancer (OC) patients; however, little is known about its prognostic value. We sought to correlate IL-10 levels in ascites and sera of OC patients with clinicopathologic characteristics and oncologic outcomes. IL-10 levels and clinical data from biobanked ascites and serum samples of OC patients were evaluated. Receiver operating characteristic curves were used to quantify marker performance and identify IL-10-high and IL-10-low groups. Correlations between IL-10 levels and clinicopathologic data were performed. Survival outcomes were calculated, while the factors affecting them were also investigated. A total of 106 patients had ascites samples, of which 44 serum samples were also available. Mean ascites IL-10 levels were significantly higher in patients with serous histology compared to endometrioid histology (p = 0.024). Fold-change in ascites IL-10 during treatment positively correlated with clinical response, as determined by a change in serum cancer antigen (CA)-125 levels (p = 0.0126). Median progression-free survival (PFS) and overall survival (OS) were shorter in patients with high compared with low ascites IL-10 levels (PFS: 18 versus 60 months; p = 0.007, OS: 42 versus 85 months; p = 0.029). A significant positive correlation was seen between ascites and sera IL-10 levels (p = 0.019). In multivariable analyses, a high ascites IL-10 level was associated with a significantly worse prognosis (PFS hazard ratio (HR) = 1.93; p = 0.02). Patients with high ascites levels of IL-10 have worse outcomes, which are likely reflective of the immunosuppressive effect of IL-10. This highlights its potential role as an immunomodulator in the tumor microenvironment, leading to OC immune evasion.
Collapse
Affiliation(s)
- Paul Adrien Guigue
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Yoav Brezinov
- Department of Experimental Surgery, McGill University, Montreal, QC H3T 1E2, Canada
| | - Amber Yasmeen
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Maroua Mbarik
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Shannon Salvador
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Susie Lau
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Walter Henri Gotlieb
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Experimental Surgery, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Melica Nourmoussavi Brodeur
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
16
|
Li W, You J, Xue H, Liu Y, Chen J, Zheng X, Chen L, Wu C. Unlocking the potential of HHLA2: identifying functional immune infiltrating cells in the tumor microenvironment and predicting clinical outcomes in laryngeal squamous cell carcinoma. Cancer Immunol Immunother 2024; 73:207. [PMID: 39105870 PMCID: PMC11303638 DOI: 10.1007/s00262-024-03791-6] [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: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND HHLA2 (human endogenous retrovirus-H long terminal repeat-associating protein 2) represents a recently identified member of the B7 immune checkpoint family, characterized by limited expression in normal tissues but notable overexpression in various cancer types. Nevertheless, the precise function and interaction with immune cells remain poorly understood, particularly in laryngeal squamous cell carcinoma (LSCC). This investigation endeavored to elucidate the biological significance of HHLA2 within the tumor microenvironment of human LSCC tissues and delineate the clinical relevance and functional roles of HHLA2 in LSCC pathogenesis. METHODS Through multiplexed immunohistochemistry analyses conducted on tissue microarrays sourced from LSCC patients (n = 72), the analysis was executed to assess the expression levels of HHLA2, density and spatial patterns of CD68+HLA-DR+CD163- (M1 macrophages), CTLA-4+CD4+FoxP3+ (CTLA-4+Treg cells), CTLA-4+CD4+FoxP3- (CTLA-4+Tcon cells), exhausted CD8+T cells, and terminally exhausted CD8+T cells in LSCC tissues. Survival analysis was conducted to evaluate the prognostic significance of HHLA2 and these immune checkpoints or immune cell populations, employing COX regression analysis to identify independent prognostic factors. RESULTS Kaplan-Meier (K-M) survival curves revealed a significant association between HHLA2 expression and overall survival (OS) in LSCC. Elevated levels of HHLA2 were linked to reduced patient survival, indicating its potential as a prognostic marker (HR: 3.230, 95%CI 0.9205-11.34, P = 0.0067). Notably, increased infiltration of CD68+ cells (total macrophages), STING+CD68+HLA-DR+CD163- (STING+M1 macrophages), CTLA-4+CD4+FoxP3+, CTLA-4+CD4+FoxP3-, PD-1+LAG-3+CD8+T cells, and PD-1+LAG-3+TIM-3+CD8+T cells strongly linked to poorer survival outcomes (P < 0.05). A discernible trend was observed between the levels of these immune cell populations, STING+CD68+ (STING+ total macrophages), CD68+HLA-DR+CD163-, STING+CD68+CD163+HLA-DR- (STING+M2 macrophages), PD-1+LAG-3-CD8+T cells, PD-1+TIM-3+CD8+T cells, and PD-1+LAG-3+TIM-3-CD8+T cells and prognosis. Importantly, multivariate COX analysis identified HHLA2 as an independent predictive factor for OS in LSCC patients (HR = 3.86, 95% CI 1.08-13.80, P = 0.038). This underscored the potential of HHLA2 as a critical marker for predicting patient outcomes in LSCC. CONCLUSIONS HHLA2 emerged as a detrimental prognostic biomarker for assessing OS in LSCC patients. Relative to other immune checkpoints, HHLA2 exhibited heightened predictive efficacy for the prognosis of LSCC patients.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Jianqing You
- Department of Otorhinolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Haixiang Xue
- Department of Otorhinolaryngology Head and Neck Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yi Liu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
17
|
Park C, Suh KJ, Kim SH, Lee KH, Im SA, Kim MH, Sohn J, Jeong JH, Jung KH, Lee KE, Park YH, Kim HJ, Cho EK, Choi IS, Noh SJ, Shin I, Cho DY, Kim JH. Genomic and transcriptomic profiles associated with response to eribulin and nivolumab combination in HER-2-negative metastatic breast cancer. Cancer Immunol Immunother 2024; 73:197. [PMID: 39105849 PMCID: PMC11303363 DOI: 10.1007/s00262-024-03782-7] [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: 06/11/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Biomarkers for predicting response to the immunotherapy and chemotherapy combination in breast cancer patients are not established. In this study, we report exploratory genomic and transcriptomic analyses of pretreatment tumor tissues from patients enrolled in phase II clinical trial of a combination of eribulin and nivolumab for HER-2-negative metastatic breast cancer (MBC) (KORNELIA trial, NCT04061863). METHODS We analyzed associations between tumor molecular profiles based on genomic (n = 76) and transcriptomic data (n = 58) and therapeutic efficacy. Patients who achieved progression-free survival (PFS) ≥ 6 months were defined as PFS6-responders and PFS6-nonresponders otherwise. FINDINGS Analyses on tumor mutation burden (TMB) showed a tendency toward a favorable effect on efficacy, while several analyses related to homologous recombination deficiency (HRD) indicated a potentially negative impact on efficacy. Patients harboring TP53 mutations showed significantly poor PFS6 rate and PFS, which correlated with the enrichment of cell cycle-related signatures in PFS6-nonresponders. High antigen presentation gene set enrichment scores (≥ median) were significantly associated with longer PFS. Naïve B-cell and plasma cell proportions were considerably higher in long responders (≥ 18 months). INTERPRETATION Genomic features including TMB, HRD, and TP53 mutations and transcriptomic features related to immune cell profiles and cell cycle may distinguish responders. Our findings provide insights for further exploring the combination regimen and its biomarkers in these tumors.
Collapse
Affiliation(s)
- Changhee Park
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro, Bundang-Gu, Seongnam, 13620, Republic of Korea
| | - Koung Jin Suh
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro, Bundang-Gu, Seongnam, 13620, Republic of Korea
| | - Se Hyun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro, Bundang-Gu, Seongnam, 13620, Republic of Korea.
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Min Hwan Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyoung Eun Lee
- Department of Hematology and Oncology, Ewha Womans University Hospital, Seoul, Korea
| | - Yeon Hee Park
- Hematology-Oncology, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee-Jun Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Eun Kyung Cho
- Division of Oncology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - In Sil Choi
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | | | | | | | - Jee Hyun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro, Bundang-Gu, Seongnam, 13620, Republic of Korea
| |
Collapse
|
18
|
Li Z, Zhang Y, Ma M, Wang W, Hui H, Tian J, Chen Y. Targeted mitigation of neointimal hyperplasia via magnetic field-directed localization of superparamagnetic iron oxide nanoparticle-labeled endothelial progenitor cells following carotid balloon catheter injury in rats. Biomed Pharmacother 2024; 177:117022. [PMID: 38917756 DOI: 10.1016/j.biopha.2024.117022] [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: 04/03/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The transplantation of endothelial progenitor cells (EPCs) has been shown to reduce neointimal hyperplasia following arterial injury. However, the efficacy of this approach is hampered by limited homing of EPCs to the injury site. Additionally, the in vivo recruitment and metabolic activity of transplanted EPCs have not been continuously monitored. METHODS EPCs were labeled with indocyanine green (ICG)-conjugated superparamagnetic iron oxide nanoparticles (SPIONs) and subjected to external magnetic field targeting to enhance their delivery to a carotid balloon injury (BI) model in Sprague-Dawley rats. Magnetic particle imaging (MPI)/ fluorescence imaging (FLI) multimodal in vivo imaging, 3D MPI/CT imaging and MPI/FLI ex vivo imaging was performed after injury. Carotid arteries were collected and analyzed for pathology and immunofluorescence staining. The paracrine effects were analyzed by enzyme-linked immunosorbent assay. RESULTS The application of a magnetic field significantly enhanced the localization and retention of SPIONs@PEG-ICG-EPCs at the site of arterial injury, as evidenced by both in vivo continuous monitoring and ex vivo by observation. This targeted delivery approach effectively inhibited neointimal hyperplasia and increased the presence of CD31-positive cells at the injury site. Moreover, serum levels of SDF-1α, VEGF, IGF-1, and TGF-β1 were significantly elevated, indicating enhanced paracrine activity. CONCLUSIONS Our findings demonstrate that external magnetic field-directed delivery of SPIONs@PEG-ICG-EPCs to areas of arterial injury can significantly enhance their therapeutic efficacy. This enhancement is likely mediated through increased paracrine signaling. These results underscore the potential of magnetically guided SPIONs@PEG-ICG-EPCs delivery as a promising strategy for treating arterial injuries.
Collapse
Affiliation(s)
- Zhongxuan Li
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingqian Zhang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Mingrui Ma
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Wang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China; National Key Laboratory of Kidney Diseases, Beijing 100853, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China; School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing 100191, China; National Key Laboratory of Kidney Diseases, Beijing 100853, China.
| | - Yundai Chen
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
19
|
Qin Y, Zhang T, Du Z, Chen S, Li Y, Lv Y, Du X, Hu Y, Liu Z. Prognosis of immune checkpoint inhibitor-related myocarditis: Retrospective experience of a single institution. Int Immunopharmacol 2024; 136:112385. [PMID: 38850788 DOI: 10.1016/j.intimp.2024.112385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/20/2024] [Accepted: 05/29/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Myocarditis related to immune checkpoint inhibitors (ICIs) treatment is a rare but potentially life-threatening adverse event. To gain insight into this condition, we analyzed the clinical characteristics and prognosis of patients with ICI-related myocarditis. METHODS Data on the clinical characteristics, management, and outcomes of patients diagnosed with ICI-related myocarditis between August 2018 and August 2023 in our institution were gathered retrospectively from medical records. Outcomes included the occurrence of major adverse cardiac events (MACE). RESULTS Among 8875 patients who received ICI therapy, 31 patients experienced ICI-related myocarditis. These 31 patients had a mean age of 62 ± 12 years and included 24 (77.4 %) males and 19 patients (61.3 %) with at least one risk factor for cardiovascular disease. The median duration from ICI initiation to the onset of myocarditis symptoms was 6.3 weeks (interquartile range, 4.3-8.1 weeks). Twenty-one patients (67.7 %) developed grade 3-4 myocarditis. Thirteen patients (42 %) experienced MACE after myocarditis onset, and 15 patients (48.4 %) showed a troponin rise > 4 times the maximum limit of the standard range. On receiver operating characteristic curve analysis, troponin level could predict MACE in patients with ICI-related myocarditis with an area under the curve of 0.82 (95 % confidence interval [CI]: 0.66-0.98, p = 0.003). From Kaplan-Meier analysis, the occurrence of MACE (p = 0.002) was an independent influencing factor on patients' overall survival. CONCLUSIONS ICI-related myocarditis frequently leads to MACE, which is associated with poor prognosis. Elevated troponin levels and electrocardiogram abnormalities in these patients may help predict the occurrence of MACE.
Collapse
Affiliation(s)
- Yuhui Qin
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Teng Zhang
- Department of Oncology, The 983rd Hospital of Joint Logistic Support Force of PLA, Tianjin, China; Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhijuan Du
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Siyuan Chen
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Yanan Li
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yahui Lv
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Xiangyu Du
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Yi Hu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| | - Zhefeng Liu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
20
|
Huang J, Zhang L, Li B, Lian Y, Lin X, Li Z, Zhang B, Feng Z. Bibliometric and visual analysis in the field of two-dimensions nano black phosphorus in cancer from 2015 to 2023. Discov Oncol 2024; 15:260. [PMID: 38961044 PMCID: PMC11222346 DOI: 10.1007/s12672-024-01104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
This study aims to provide a comprehensive summary of the status and trends of Two-Dimensional Nano Black Phosphorus (2D nano BP) in cancer research from 2015 to 2023, offering insights for future studies. To achieve this, articles from the Web of Science database published between 2015 and 2023 were analyzed using R and VOSviewer software. The analysis included 446 articles, revealing a consistent increase in publication rates, especially between 2017 and 2019. China emerged as a leader in both publication volume and international collaborations. Prominent journals in this field included ACS Applied Materials & Interfaces and Advanced Materials, while key researchers were identified as Zhang Han, Tao Wei, and Yu Xuefeng. The analysis highlighted common keywords such as drug delivery, photothermal therapy, photodynamic therapy, and immunotherapy, indicating the major research focuses. The findings suggest that 2D nano BP holds significant promise in cancer treatment research, with a growing global interest. This study thus serves as a valuable reference for future investigations, providing a detailed analysis of the current state and emerging trends in this promising field.
Collapse
Affiliation(s)
- Jing'an Huang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Ling Zhang
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Boren Li
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yuanchu Lian
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Xiaoxin Lin
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Zonghuai Li
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China.
| | - Zhongwen Feng
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|
21
|
Ramapriyan R, Vykunta VS, Vandecandelaere G, Richardson LGK, Sun J, Curry WT, Choi BD. Altered cancer metabolism and implications for next-generation CAR T-cell therapies. Pharmacol Ther 2024; 259:108667. [PMID: 38763321 DOI: 10.1016/j.pharmthera.2024.108667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
This review critically examines the evolving landscape of chimeric antigen receptor (CAR) T-cell therapy in treating solid tumors, with a particular focus on the metabolic challenges within the tumor microenvironment. CAR T-cell therapy has demonstrated remarkable success in hematologic malignancies, yet its efficacy in solid tumors remains limited. A significant barrier is the hostile milieu of the tumor microenvironment, which impairs CAR T-cell survival and function. This review delves into the metabolic adaptations of cancer cells and their impact on immune cells, highlighting the competition for nutrients and the accumulation of immunosuppressive metabolites. It also explores emerging strategies to enhance CAR T-cell metabolic fitness and persistence, including genetic engineering and metabolic reprogramming. An integrated approach, combining metabolic interventions with CAR T-cell therapy, has the potential to overcome these constraints and improve therapeutic outcomes in solid tumors.
Collapse
Affiliation(s)
- Rishab Ramapriyan
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Vivasvan S Vykunta
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gust Vandecandelaere
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leland G K Richardson
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jing Sun
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - William T Curry
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bryan D Choi
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
22
|
Ahmadpour S, Habibi MA, Ghazi FS, Molazadeh M, Pashaie MR, Mohammadpour Y. The effects of tumor-derived supernatants (TDS) on cancer cell progression: A review and update on carcinogenesis and immunotherapy. Cancer Treat Res Commun 2024; 40:100823. [PMID: 38875884 DOI: 10.1016/j.ctarc.2024.100823] [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: 04/25/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/16/2024]
Abstract
Tumors can produce bioactive substances called tumor-derived supernatants (TDS) that modify the immune response in the host body. This can result in immunosuppressive effects that promote the growth and spread of cancer. During tumorigenesis, the exudation of these substances can disrupt the function of immune sentinels in the host and reinforce the support for cancer cell growth. Tumor cells produce cytokines, growth factors, and proteins, which contribute to the progression of the tumor and the formation of premetastatic niches. By understanding how cancer cells influence the host immune system through the secretion of these factors, we can gain new insights into cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Sajjad Ahmadpour
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mikaeil Molazadeh
- Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Pashaie
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Mohammadpour
- Department of Medical Education, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
23
|
Zou R, Hao Y, Qi C, Peng X, Huang Z, Li D, Wang Y. Trimethyl chitosan-cysteine-based nanoparticles as an effective delivery system for portulacerebroside A in the management of hepatocellular carcinoma cells in vitro and in vivo. J Drug Target 2024; 32:570-584. [PMID: 38625591 DOI: 10.1080/1061186x.2024.2344495] [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: 12/14/2023] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
Portulacerebroside A (PCA), a cerebroside compound extracted from Portulaca oleracea L., has been shown to suppress hepatocellular carcinoma (HCC) cells. This study aims to investigate the effectiveness of trimethyl chitosan-cysteine (TMC-Cys) nanocarrier in delivering PCA for HCC management and to elucidate the molecular mechanisms behind PCA's function. TMC-Cys nanocarriers notably augmented PCA's function, diminishing the proliferation, migration, and invasiveness of HCC cells in vitro, reducing hepatocellular tumorigenesis in immunocompetent mice, and impeding metastasis of xenograft tumours in nude mice. Comprehensive bioinformatics analyses, incorporating Super-PRED systems alongside pathway enrichment analysis, pinpointed toll-like receptor 4 (TLR4) and epidermal growth factor receptor (EGFR) as two promising targets of PCA, enriched in immune checkpoint pathway. PCA/nanocarrier (PCA) reduced levels of TLR4 and EGFR and their downstream proteins, including programmed cell death ligand 1, thereby increasing populations and activity of T cells co-cultured with HCC cells in vitro or in primary HCC tumours in mice. However, these effects were counteracted by additional artificial activation of TLR4 and EGFR. In conclusion, this study provides novel evidence of PCA's function in immunomodulation in addition to its direct tumour suppressive effect. TMC-Cys nanocarriers significantly enhance PCA efficacy, indicating promising application as a drug delivery system.
Collapse
Affiliation(s)
- Rui Zou
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, Haikou, P.R. China
| | - Yunhe Hao
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, Haikou, P.R. China
| | - Chunchun Qi
- Medical College of Nankai University, Tianjin, P.R. China
| | - Xu Peng
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, Haikou, P.R. China
| | - Zepeng Huang
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, Haikou, P.R. China
| | - Duo Li
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, Haikou, P.R. China
| | - Yiyao Wang
- Department of Integrated Traditional Chinese and Western Medicine, Hainan Cancer Hospital, Haikou, P.R. China
| |
Collapse
|
24
|
Wei Y, Guo J, Meng T, Gao T, Mai Y, Zuo W, Yang J. The potential application of complement inhibitors-loaded nanosystem for autoimmune diseases via regulation immune balance. J Drug Target 2024; 32:485-498. [PMID: 38491993 DOI: 10.1080/1061186x.2024.2332730] [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: 12/20/2023] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
The complement is an important arm of the innate immune system, once activated, the complement system rapidly generates large quantities of protein fragments that are potent mediators of inflammation. Recent studies have shown that over-activated complement is the main proinflammatory system of autoimmune diseases (ADs). In addition, activated complements interact with autoantibodies, immune cells exacerbate inflammation, further worsening ADs. With the increasing threat of ADs to human health, complement-based immunotherapy has attracted wide attention. Nevertheless, efficient and targeted delivery of complement inhibitors remains a significant challenge owing to their inherent poor targeting, degradability, and low bioavailability. Nanosystems offer innovative solutions to surmount these obstacles and amplify the potency of complement inhibitors. This prime aim to present the current knowledge of complement in ADs, analyse the function of complement in the pathogenesis and treatment of ADs, we underscore the current situation of nanosystems assisting complement inhibitors in the treatment of ADs. Considering technological, physiological, and clinical validation challenges, we critically appraise the challenges for successfully translating the findings of preclinical studies of these nanosystem assisted-complement inhibitors into the clinic, and future perspectives were also summarised. (The graphical abstract is by BioRender.).
Collapse
Affiliation(s)
- Yaya Wei
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jueshuo Guo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Tingting Meng
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ting Gao
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yaping Mai
- School of Science and Technology Centers, Ningxia Medical University, Yinchuan, China
| | - Wenbao Zuo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
25
|
Chen G, Xiong M, Jiang C, Zhao Y, Chen L, Ju Y, Jiang J, Xu Z, Pan J, Li X, Wang K. Novel BODIPY-based nano-biomaterials with enhanced D-A-D structure for NIR-triggered photodynamic and photothermal therapy. Bioorg Chem 2024; 148:107494. [PMID: 38797067 DOI: 10.1016/j.bioorg.2024.107494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Near-infrared (NIR) responsive nanoparticles are an important platform for multimodal phototherapy. Importantly, the simultaneous NIR-triggered photodynamic (PDT) and photothermal (PTT) therapy is a powerful approach to increase the antitumor efficiency of phototherapic nanoparticles due to the synergistic effect. Herein, a boron dipyrromethene (BODIPY)-based amphiphilic dye with enhanced electron donor-acceptor-donor (D-A-D) structure (BDP-AP) was designed and synthesized, which could self-assemble into stable nanoparticles (BDP-AP NPs) for the synergistic NIR-triggered PDT/PTT therapy. BDP-AP NPs synchronously generated singlet oxygen (1O2) and achieved preeminent photothermal conversion efficiency (61.42%). The in vitro and in vivo experiments showed that BDP-AP NPs possessed negligible dark cytotoxicity and infusive anticancer performance. BDP-AP NPs provide valuable guidance for the construction of PDT/PTT-synergistic NIR nanoagents to improve the efficiency of photoinduced cancer therapy in the future.
Collapse
Affiliation(s)
- Gang Chen
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Mengmeng Xiong
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Chen Jiang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Yimei Zhao
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China.
| | - Li Chen
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Yunlong Ju
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Jun Jiang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China; Hubei Province Engineering Centre of Performance Chemicals, Wuhan 430062, PR China.
| | - Zekun Xu
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Jie Pan
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China
| | - Xiang Li
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China.
| | - Kai Wang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China.
| |
Collapse
|
26
|
Yuan M, Han Z, Li Y, Zhan X, Sun Y, He B, Liang Y, Luo K, Li F. A pH-responsive nanoplatform with dual-modality imaging for enhanced cancer phototherapy and diagnosis of lung metastasis. J Nanobiotechnology 2024; 22:180. [PMID: 38622591 PMCID: PMC11017640 DOI: 10.1186/s12951-024-02431-6] [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: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024] Open
Abstract
To address the limitations of traditional photothermal therapy (PTT)/ photodynamic therapy (PDT) and real-time cancer metastasis detection, a pH-responsive nanoplatform (NP) with dual-modality imaging capability was rationally designed. Herein, 1 H,1 H-undecafluorohexylamine (PFC), served as both an oxygen carrier and a 19F magnetic resonance imaging (MRI) probe, and photosensitizer indocyanine green (ICG) were grafted onto the pH-responsive peptide hexahistidine (H6) to form H6-PFC-ICG (HPI). Subsequently, the heat shock protein 90 inhibitor, gambogic acid (GA), was incorporated into hyaluronic acid (HA) modified HPI (HHPI), yielding the ultimate HHPI@GA NPs. Upon self-assembly, HHPI@GA NPs passively accumulated in tumor tissues, facilitating oxygen release and HA-mediated cell uptake. Once phagocytosed by lysosomes, protonation of H6 was triggered due to the low pH, resulting in the release of GA. With near-infrared laser irradiation, GA-mediated decreased HSP90 expression and PFC-mediated increased ROS generation amplified the PTT/PDT effect of HHPI@GA, leading to excellent in vitro and in vivo anticancer efficacies. Additionally, the fluorescence and 19F MRI dual-imaging capabilities of HHPI@GA NPs enabled effective real-time primary cancer and lung metastasis monitoring. This work offers a novel approach for enhanced cancer phototherapy, as well as precise cancer diagnosis.
Collapse
Affiliation(s)
- Mujie Yuan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Zeyu Han
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xin Zhan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yong Sun
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Yan Liang
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China.
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fan Li
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China.
| |
Collapse
|
27
|
Pant A, Singh G, Barnwal RP, Sharma T, Singh B. QbD-driven development and characterization of superparamagnetic iron oxide nanoparticles (SPIONS) of a bone-targeting peptide for early detection of osteoporosis. Int J Pharm 2024; 654:123936. [PMID: 38417727 DOI: 10.1016/j.ijpharm.2024.123936] [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: 12/17/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
Osteoporosis is a metabolic disorder that leads to deterioration of bones. The major challenges confronting osteoporosis therapy include early-stage detection and regular disease monitoring. The present studies employed D-aspartic acid octapeptide (-D-Asp-)8 as bone-targeting peptide for evaluating osteoporosis manifestation, and superparamagnetic iron oxide nanoparticles (SPIONs) as nanocarriers for MRI-aided diagnosis. Thermal decomposition technique was employed to synthesize SPIONs, followed by surface-functionalization with hydrophilic ligands. Failure mode effect analysis and factor screening studies were performed to identify concentrations of SPIONs and ligand as critical material attributes, and systematic optimization was subsequently conducted employing face-centered cubic design. The optimum formulation was delineated using desirability function, and design space demarcated with 178.70 nm as hydrodynamic particle size, -24.40 mV as zeta potential, and 99.89 % as hydrophilic iron content as critical quality attributes. XRD patterns ratified lattice structure and SQUID studies corroborated superparamagnetic properties of hydrophilic SPIONs. Bioconjugation of (-D-Asp-)8 with SPIONs (1:1) was confirmed using UV spectroscopy, FTIR and NMR studies. Cell line studies indicated successful targeting of SPIONs to MG-63 human osteoblasts, ratifying enormous bone-targeting and safety potential of peptide-tethered SPIONs as MRI probes. In vivo MRI imaging studies in rats showcased promising contrast ability and safety of peptide-conjugated SPIONs.
Collapse
Affiliation(s)
- Anjali Pant
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India.
| | | | - Teenu Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140 401, India
| | - Bhupinder Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India; Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140 401, India.
| |
Collapse
|
28
|
Li L, Le C, Liu W, Zhang H, Chen X, Yang J, Fang M, Zhang X. Toll-like receptor 2 deficiency promotes the generation of alloreactive γδT17 cells after cardiac transplantation in mice. Int Immunopharmacol 2024; 130:111768. [PMID: 38432149 DOI: 10.1016/j.intimp.2024.111768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Homograft rejection is the main cause of heart transplantation failure. The role of TLR2, a major member of the toll-like receptor (TLR) family, in transplantation rejection is has yet to be elucidated. In this study, we used a mouse model of acute cardiac transplantation rejection to investigate whether the TLR2 signalling pathway can regulate cardiac transplantation rejection by regulating alloreactive IL-17+γδT (γδT17) cells. We found that the expression of TLR2 on the surface of dendritic cells (DCs) and macrophages increased during acute transplantation rejection. In addition, our investigation revealed that γδT17 cells exert a significant influence on acute cardiac transplantation rejection. TLR2 gene knockout resulted in an increase in alloreactive γδT17 cells in the spleen and heart grafts of recipient mice compared with wild-type recipient mice and an increase in the mRNA expression of IL-17, IL-1β, CCR6, and CCL20 in the heart grafts. In an in vitro experiment, a mixed lymphocyte reaction was conducted to assess the impact of TLR2 deficiency on the generation of γδT17 cells, which further substantiated a significant increase compared to that in wild-type controls. Furthermore, the mixed lymphocyte reaction showed that TLR2 regulated the production of γδT17 cells by regulating the ability of DCs to secrete IL-1β. These results suggest that TLR2 signalling is important for regulating the generation of γδT17 cells after cardiac allograft transplantation.
Collapse
Affiliation(s)
- Lingyun Li
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Chenyu Le
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Wenfei Liu
- Chinese PLA 305 Hospital, Beijing 100018, China
| | - Hongchen Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Xiangyu Chen
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China; The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Min Fang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China; The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310006, China; Hangzhou Institute of Digestive Disease, Hangzhou 310006, China.
| |
Collapse
|
29
|
Sun C, Zhang Y, Wang Z, Chen J, Zhang J, Gu Y. TMED2 promotes glioma tumorigenesis by being involved in EGFR recycling transport. Int J Biol Macromol 2024; 262:130055. [PMID: 38354922 DOI: 10.1016/j.ijbiomac.2024.130055] [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: 11/02/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Aberrant epidermal growth factor receptor (EGFR) signaling is the core signaling commonly activated in glioma. The transmembrane emp24 protein transport domain protein 2 (TMED2) interacts with cargo proteins involved in protein sorting and transport between endoplasmic reticulum (ER) and Golgi apparatus. In this study, we found the correlation between TMED2 with glioma progression and EGFR signaling through database analysis. Moreover, we demonstrated that TMED2 is essential for glioma cell proliferation, migration, and invasion at the cellular levels, as well as tumor formation in mouse models, underscoring its significance in the pathobiology of gliomas. Mechanistically, TMED2 was found to enhance EGFR-AKT signaling by facilitating EGFR recycling, thereby providing the initial evidence of TMED2's involvement in the membrane protein recycling process. In summary, our findings shed light on the roles and underlying mechanisms of TMED2 in the regulation of glioma tumorigenesis and EGFR signaling, suggesting that targeting TMED2 could emerge as a promising therapeutic strategy for gliomas and other tumors associated with aberrant EGFR signaling.
Collapse
Affiliation(s)
- Changning Sun
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China
| | - Yihan Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China
| | - Zhuangzhi Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China
| | - Jin Chen
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Junhua Zhang
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Laboratory for Marine Drugs and Bioproducts of Laoshan Laboratory, Qingdao 266200, China.
| |
Collapse
|
30
|
Magisetty J, Gadiraju B, Kondreddy V. Genomic analysis in the colon tissues of omega-3 fatty acid-treated rats identifies novel gene signatures implicated in ulcerative colitis. Int J Biol Macromol 2024; 258:128867. [PMID: 38123036 DOI: 10.1016/j.ijbiomac.2023.128867] [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: 08/14/2023] [Revised: 11/11/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Several long-term intervention trials only studied the ex vivo immunological function to elucidate the beneficial mechanisms of n-3 polyunsaturated fatty acids (PUFA) in the ulcerative colitis (UC). An unbiased whole-transcriptome analysis would be more valuable to obtain a comprehensive understanding of the processes and genes regulated by n-3 PUFA in vivo. In this study, we have performed microarray analysis in the colon tissues of dextran sulfate sodium (DSS)-induced UC in rats supplemented with n-6 PUFA, n-3PUFA and long-chain n-3PUFA (LC-n3PUFA). We have identified the novel gene signatures previously not linked to colitis such as Etv3, Clec4d, CD180, CD72, Megf11, and Angptl4 which are most downregulated in both n-3PUFA and LC-n3PUFA groups compared to the n-6PUFA group. The most upregulated genes were Nr1i3, Nptx2, and Zfp810 in both n-3PUFA and LC-n3PUFA groups. The RT-PCR analysis confirmed similar results. Interestingly, LPS treatment in macrophages upregulated the Megf11, Etv3, CD180, and Angptl4, and correlated with increased secretion of cytokines. Gene silencing of Etv3, Megf11, and CD180 in rats using intravascular delivery of siRNA-lipoparticles attenuated the DSS-induced ulceration and mucosal damage. Thus, our genome-wide microarray analysis identified novel genes regulated by omega-3 PUFA and offers new drug targets that could prevent or reduce UC.
Collapse
Affiliation(s)
- Jhansi Magisetty
- Department of Biochemistry, Central Food Technological Research Institute, Mysore 570020, India
| | - Bhavani Gadiraju
- Center for Lipid Science & Technology, The Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India
| | - Vijay Kondreddy
- Center for Lipid Science & Technology, The Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India.
| |
Collapse
|
31
|
Chen S, Chen E, Guan X, Li J, Qin A, Wang C, Fu X, Huang C, Li J, Tang Y, Wei M, Zhang L, Su J. Magnetically controlled nanorobots induced oriented and rapid clearance of the cytokine storm for acute lung injury therapy. Colloids Surf B Biointerfaces 2024; 234:113731. [PMID: 38184944 DOI: 10.1016/j.colsurfb.2023.113731] [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: 11/11/2023] [Revised: 12/19/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024]
Abstract
Cytokine storms characterized by excessive secretion of circulating cytokines and immune-cell hyperactivation are life-threatening systemic inflammatory syndromes. The new strategy is in great demand to inhibit the cytokine storm. Here, we designed a type of magnetically controlled nanorobots (MAGICIAN) by fusing neutrophil membranes onto Fe3O4 nanoparticles (Fe3O4NPs). In our study, the receptors of neutrophil membranes were successfully coated to the surface of Fe3O4NPs. The associated membrane functions of neutrophils were highly preserved. MAGICIAN could in vitro neutralize the inflammatory cytokines including interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), and interferon γ (IFN-γ). Interestingly, MAGICIAN could be navigated to the liver sites under magnetic control and accelerated the cytokine clearance by the liver. Administration of MAGICIAN could efficiently relieve the inflammation in the acute lung injury mouse model. In addition, MAGICIAN displayed good biosafety in systemic administration. The present study provides a safe and convenient approach for the clearance of cytokine storms, indicating the potential for clinical application in acute lung injury therapy.
Collapse
Affiliation(s)
- Sheng Chen
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Enen Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaoling Guan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Junfang Li
- Shimen Second Road Community Health Service Center, Jing-An District, Shanghai 200041, China
| | - Aiping Qin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Chen Wang
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China
| | - Xihua Fu
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China
| | - Chen Huang
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China
| | - Jianhao Li
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China
| | - Yukuan Tang
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China
| | - Minyan Wei
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lingmin Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Jianfen Su
- Pharmacy Department, Infection Medicine Research Institute of Panyu District, Cardiovascular Diseases Research Institute of Panyu District, Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Guangzhou 511400, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
32
|
Chen Z, Kong Y, Huang Z, Zheng X, Zheng Z, Yao D, Yang S, Zhang Y, Aweya JJ. Exogenous alpha-linolenic acid and Vibrio parahaemolyticus induce EPA and DHA levels mediated by delta-6 desaturase to enhance shrimp immunity. Int J Biol Macromol 2024; 257:128583. [PMID: 38056755 DOI: 10.1016/j.ijbiomac.2023.128583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/18/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Globally, penaeid shrimp are the most farmed and traded aquatic organisms, although they are easily susceptible to microbial pathogens. Moreover, there is a desire to increase the nutritional value of shrimp, especially the levels of n-3 polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which also possess immunomodulatory and anti-inflammatory properties. Some aquatic animals can synthesize EPA and DHA from dietary plant-sourced alpha-linolenic acid (ALA), but penaeid shrimps' ability to synthesize these n-3 PUFAs is unknown. Here, molecular biology techniques, including gas chromatography-mass spectrometry, qPCR, ELISA, etc., were used to demonstrate that exogenous ALA or Vibrio parahaemolyticus could modulate EPA and DHA levels and immune genes in Penaeus vannamei by inducing key enzymes involved in n-3 PUFAs biosynthesis, such as delta desaturases and elongation of very long-chain fatty acid (ELOVLs). Most importantly, knockdown or inhibition of ∆6 desaturase significantly decreased EPA and DHA levels and immune gene expression even with exogenous ALA treatment, consequently affecting shrimp antibacterial immunity and survival. This study provides new insight into the potential of P. vannamei to synthesize n-3 PUFAs from exogenous ALA or upon bacteria challenge, which could be leveraged to increase their nutritional content and antimicrobial immunity.
Collapse
Affiliation(s)
- Zeyan Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Yonghui Kong
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Zishu Huang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Zhihong Zheng
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Defu Yao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Shen Yang
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China.
| | - Jude Juventus Aweya
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China.
| |
Collapse
|
33
|
Zhang Z, Yuan Y, Xue Y, Zhang W, Sun X, Xu X, Liu C. Nanomaterials for Ultrasound Imaging- Guided Sonodynamic Therapy. Technol Cancer Res Treat 2024; 23:15330338241263197. [PMID: 39051705 PMCID: PMC11273702 DOI: 10.1177/15330338241263197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 07/27/2024] Open
Abstract
Ultrasound examination is becoming the most popular medical imaging modality because of its low cost and high safety profile. Ultrasound contrast agents enhance the scattering of sound waves, which can improve the clarity and resolution of images. Nanoparticle Ultrasound contrast agents have the characteristics of a large specific surface area and a modifiable surface, which can increase drug loading capacity, prolong circulation time, and enable drug enrichment in specific organs or tissues. This leads to improved therapeutic effects and reducing toxic and side effects. Compared with traditional ultrasound contrast agents, Nano-ultrasound contrast agents overcome the limitation of imaging solely within blood vessels and facilitate imaging within tumor tissues, thereby extending the duration of enhanced imaging. Sonodynamic therapy is an emerging treatment method that has been developed rapidly in recent years, which has the advantages of noninvasive, high spatial and temporal resolution, and low toxicity and side effects. Sonodynamic therapy utilizes a sonosensitizer that, when excited by ultrasound at the tumor site, produces toxic reactive oxygen species, inducing apoptosis or necrosis in tumor cells. Ultrasound-guided sonodynamic therapy allows for real-time observation of lesions, is convenient and flexible, and is free of radiation exposure. With the use of nanomaterials as carriers, ultrasound-guided sonodynamic therapy has made significant strides. This study categorizes and summarizes the current research on acoustic sensitizer carrier materials, including carbon-based, silicon-based, peptide-based, iron-based, metal-organic frameworks, polymers, and liposomes. It concludes by highlighting the current challenges in the integration of ultrasound imaging with sonodynamic therapy and suggests future directions for clinical application development.
Collapse
Affiliation(s)
- Zhiyang Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yinuo Yuan
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanzhang Xue
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenjing Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao Sun
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, China
| | - Cun Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
34
|
Li N, Wang Y, Yang L, Yang C, Zeng M. Hepatobiliary-Phase Hypointense Nodules Without Arterial-Phase Hyperenhancement: Developing a Risk Stratification for Hypervascular Transformation Based on a Real-World Observational Cohort Study. Technol Cancer Res Treat 2024; 23:15330338241299003. [PMID: 39544080 PMCID: PMC11565688 DOI: 10.1177/15330338241299003] [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: 07/14/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
PURPOSE To develop a risk stratification based on MRI features to predict hypervascular transformation for hepatobiliary-phase (HBP) hypointense nodules without arterial-phase hyperenhancement (APHE). MATERIALS AND METHODS This retrospective observational cohort study included 55 HBP hypointense nodules without APHE in 35 patients with chronic liver disease, cirrhosis, or current hepatocellular carcinoma (HCC) who underwent gadoxetic acid-enhanced MRI. The hypervascular transformation during the follow-up MRI(s) was the primary endpoint analyzed for the nodules. Univariable and multivariable Cox proportional hazard regression analyses were performed to identify risk features predicting transformation and assess their predictive value. RESULTS Among the 55 nodules, 27 developed hypervascular transformation, while 28 did not. Diffusion-weighted imaging (DWI) hyperintensity (hazard ratio [HR], 4.98; 95% confidence interval [CI]: 1.60, 15.54; p = 0.006) and portal venous phase (PVP) hypointensity (HR, 4.08; 95% CI: 1.43, 11.64; p = 0.009) were associated with hypervascular transformation. DWI hyperintensity and PVP hypointensity had 44.4% (95% CI: 26.0%, 64.4%) and 81.9% (95% CI: 61.3%, 93.0%) sensitivity, while their specificity was 78.2% (95% CI: 64.6%, 87.8%) and 67.9 (95% CI: 47.6%, 83.4%), respectively. The specificity of the combination of two features was 100% (95% CI: 85.0%, 100%). The hypervascular transformation rates for nodules with both, either and neither of the risk MRI findings were 100% (10/10), 60.9% (14/23), and 13.6% (3/22), respectively; the median intervals for transformation were 312 (range: 73-838), 409 (range: 50-1643) and 555 (range: 423-968) days, respectively. CONCLUSION The combination of DWI hyperintensity and PVP hypointensity may be used as a high-risk indicator for the hypervascular transformation of HBP hypointense nodules without APHE; nodules without either feature may be treated as low-risk nodules and could adopt an extended interval follow-up schedule.
Collapse
Affiliation(s)
- Na Li
- Shanghai Institute of Medical Imaging, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Wang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Yang
- Shanghai Institute of Medical Imaging, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun Yang
- Shanghai Institute of Medical Imaging, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengsu Zeng
- Shanghai Institute of Medical Imaging, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Medical Imaging, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Cong Y, Zhang SY, Li HM, Zhong JJ, Zhao W, Tang YJ. A truncated DNA aptamer with high selectivity for estrogen receptor-positive breast cancer cells. Int J Biol Macromol 2023; 252:126450. [PMID: 37634779 DOI: 10.1016/j.ijbiomac.2023.126450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023]
Abstract
The estrogen receptor-positive (ER+) breast cancers constitute more than 50 % of breast cancers, seriously threatening the health of women. Unfortunately, the detection and targeted therapy of ER+ breast cancers remain a challenge. Here, a novel nucleic acid aptamer S1-4 was developed to specifically target ER+ breast cancer MCF-7 cells by using Cell-SELEX and nucleic acid truncation strategies. The affinity dissociation constant of the binding of aptamer S1-4 to MCF-7 cells was 97.6 ± 7.5 nM in vitro. Compared with HER2+ breast cells SK-BR-3 and triple-negative breast cancer cells MDA-MB-231, MCF-7 cells were selectively recognized and targeted by aptamer S1-4. Fluorescence tracing in vivo results also indicated that aptamer S1-4 selectively targeted the cell membrane of tumor tissues in MCF-7- but not in SK-BR3 or MDB-MA-231-bearing mice. This selectively developed novel aptamer probe S1-4 with high affinity could be used for the diagnosis and treatment of ER+ breast cancers.
Collapse
Affiliation(s)
- Ying Cong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Shu-Yue Zhang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| |
Collapse
|
36
|
Vartak R, Deore B, Sanhueza CA, Patel K. Cetuximab-based PROteolysis targeting chimera for effectual downregulation of NSCLC with varied EGFR mutations. Int J Biol Macromol 2023; 252:126413. [PMID: 37598823 DOI: 10.1016/j.ijbiomac.2023.126413] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
PROteolysis Targeting Chimeras (PROTACs) showed tremendous therapeutic potential in degrading several oncoproteins including undruggable proteins. PROTACs are bifunctional molecules where one-part binds to target protein while the other end recruits protein degradation machinery. With the unveiling advancements in the field of PROTACs, we explored a combinatorial approach by developing antibody-based PROTAC (ABTAC) which may effectively degrade one of the key oncoprotein driving proliferation and progression of cancer - Epidermal growth factor receptor (EGFR). The objective of current research was to synthesize and characterize an EGFR degrading ABTAC for the treatment of non-small cell lung cancer (NSCLC). Cetuximab and pomalidomide (E3 ligase recruiting ligand) were conjugated using lysine conjugation and copper free azide-alkyne cycloaddition (CuAAC) click chemistry. Analytical characterization using reverse-phase liquid chromatography and mass spectrometry suggested conjugation of five E3-ligase inhibitor molecules/antibody. Nearly 10-30 folds reduction in IC50 was observed with ABTAC in HCC827 (EGFR sensitive) and H1650 (EGFR resistant) cells compared to cetuximab. Multicellular 3D spheroid assay strongly suggested that ABTAC induced significant apoptosis and also inhibited cell proliferation compared to control and antibody alone. Circular dichroism and surface plasmon resonance (SPR) confirmed minor alterations in the structure and receptor binding efficacy of the antibody post-conjugation.
Collapse
Affiliation(s)
- Richa Vartak
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States of America
| | - Bhavesh Deore
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States of America
| | - Carlos A Sanhueza
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States of America
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States of America.
| |
Collapse
|
37
|
Jiang W, Lei Y, Peng C, Wu D, Wu J, Xu Y, Xia X. Recent advances in cancer cell bionic nanoparticles for tumour therapy. J Drug Target 2023; 31:1065-1080. [PMID: 37962304 DOI: 10.1080/1061186x.2023.2283838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
Nanoparticle-based drug delivery systems have found extensive use in delivering oncology therapeutics; however, some delivery vehicles still exhibit rapid immune clearance, lack of biocompatibility and insufficient targeting. In recent years, bionanoparticles constructed from tumour cell membranes have gained momentum as tumour-targeting therapeutic agents. Cancer cell membrane-coated nanoparticles (CCMCNPs) typically consist of a drug-loaded nanoparticle core coated with cancer cell membrane. CCMCNPs retain homologous tumour cell surface antigens, receptors and proteins, and it has been shown that the modified nanoparticles exhibit better homologous targeting, immune escape and biocompatibility. CCMCNPs are now widely used in a variety of cancer treatments, including photothermal, photodynamic and sonodynamic therapies, chemotherapy, immunotherapy, chemodynamical therapy or other combination therapies. This article presents different therapeutic approaches using multimodal antitumour therapy-combination of two or more therapies that treat tumours synergistically-based on tumour cell membrane systems. The advantages of CCMCNPs in different cancer treatments in recent years are summarised, thus, providing new strategies for cancer treatment research.
Collapse
Affiliation(s)
- Wanting Jiang
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yujing Lei
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Cheng Peng
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Donghai Wu
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Wu
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yiling Xu
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Xinhua Xia
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
38
|
Sokkar MF, Hamdy M, Erian PS, Mosaad RM, Elaraby NM, Taher MB, El-Sayed H, Al Komy M, Eid MM, Mohamed AM, Amr KS, El-Kamah GY. Studying the pathogenicity of 26 variants characterized in the first molecular analyses of Egyptian aplastic anemia patients. J Genet Eng Biotechnol 2023; 21:149. [PMID: 38017244 PMCID: PMC10684839 DOI: 10.1186/s43141-023-00585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Aplastic anemia (AA) is a bone marrow disorder characterized by peripheral pancytopenia and marrow hypoplasia which can lead to life-threatening complications. Our objective was to study the telomerase genes (TERT and TERC) variants, explore their relationship to telomere shortening and TERT gene expression, and to identify variants in the MPL gene within Egyptian AA patients. METHODS Forty AA patients and 40 sex- and age-matched healthy individuals as the control group were studied through sequencing of TERT, TERC, and MPL genes. Quantitative real-time PCR (qRT-PCR) was used for measuring TERT gene expression. Telomere length (TL) was measured using the Quantitative Fluorescence In Situ Hybridization (Q-FISH) technique. In silico analysis was performed for the prediction of the pathogenicity of resultant variants. RESULTS Sequencing of MPL, TERT, and TERC genes identified 26 variants. Eleven variants were identified in the MPL gene. Three of them are pathogenic: two missense [c.305 G>A, c.1589 C>T] and one splice site [g.9130T>G]. TERT gene sequencing showed thirteen variants, among them, four novel [c.484G>A, c.499G>A, c.512G>A, c.3164C>G] and two previously reported [c.835G>A, c.2031C>T] were predicted to be pathogenic. Two variants were characterized within the TERC gene; n.514A>G and n.463 C>T. TERT gene expression was downregulated in 70% of studied patients and the Q-FISH technique detected telomere shortening in 82.5% of patients. CONCLUSIONS Twenty-six pathogenic and benign variants within the TERC, TERT, and MPL genes were identified among the studied AA patients that were in several cases associated with shortened telomeres and/or lower TERT gene expression. Genotype/phenotype correlation in AA patients is of great importance in explaining the disease severity and guiding therapeutic decisions.
Collapse
Affiliation(s)
- Mona F Sokkar
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt.
| | - Mona Hamdy
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Peter Sf Erian
- Human Cytogenetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Rehab M Mosaad
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Nesma M Elaraby
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Mohamed B Taher
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Heba El-Sayed
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Mohammed Al Komy
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha M Eid
- Human Cytogenetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Amal M Mohamed
- Human Cytogenetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Khalda S Amr
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Ghada Y El-Kamah
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| |
Collapse
|
39
|
Wang H, Bi X, Zhang R, Yuan H, Xu J, Zhang K, Qi S, Zhang X, Jiang M. Adipose-Derived Mesenchymal Stem Cell Facilitate Hematopoietic Stem Cell Proliferation via the Jagged-1/Notch-1/Hes Signaling Pathway. Stem Cells Int 2023; 2023:1068405. [PMID: 38020206 PMCID: PMC10653966 DOI: 10.1155/2023/1068405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/29/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Poor graft function (PGF) is a life-threatening complication following hematopoietic stem cell transplantation (HSCT). Current therapies, such as CD34+ cell infusion, have shown limited effectiveness. Conversely, mesenchymal stem cells (MSCs) show potential in addressing PGF. Adipose-derived mesenchymal stem cells (ADSCs) effectively support long-term hematopoietic stem cell proliferation. Therefore, this study aimed to investigate the mechanisms underlying the long-term hematopoietic support provided by ADSCs. Methods ADSCs were isolated from mice and subsequently identified. In vitro experiments involved coculturing ADSCs as feeders with Lin-Sca-1+c-kit+ (LSK) cells from mice for 2 and 5 weeks. The number of LSK cells was quantified after coculture. Scanning electron microscopy was utilized to observe the interaction between ADSCs and LSK cells. Hes-1 expression was assessed using western blot and real-time quantitative PCR. An γ-secretase inhibitor (GSI) was used to confirm the involvement of the Jagged-1/Notch-1/Hes-1 pathway in LSK cell expansion. Additionally, Jagged-1 was knocked down in ADSCs to demonstrate its significance in ADSC-mediated hematopoietic support. In vivo experiments were conducted to study the hematopoietic support provided by ADSCs through the infusion of LSK, LSK + fibroblasts, and LSK + ADSCs, respectively. Mouse survival, platelet count, leukocyte count, and hemoglobin levels were monitored. Results ADSCs showed high-Jagged-1 expression and promoted LSK cell proliferation. There was a direct interaction between ADSCs and LSK cells. After coculture, Hes-1 expression increased in LSK cells. Moreover, GSI-reduced LSK cell proliferation and Hes-1 expression. Knockdown of Jagged-1 attenuated ADSCs-mediated promotion of LSK cell proliferation. Furthermore, ADSCs facilitated hematopoietic recovery and promoted the survival of NOD/SCID mice. Conclusion The hematopoietic support provided by ADSCs both in vivo and in vitro may be mediated, at least in part, through the Jagged-1/Notch-1 signaling pathway. These findings provide valuable insights into the mechanisms underlying ADSCs-mediated hematopoietic support and may have implications for improving the treatment of PGF following HSCT.
Collapse
Affiliation(s)
- Hongbo Wang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Xiaojuan Bi
- The State Key Laboratory of Pathogenesis and Prevention of Central Asian High Incidence Diseases, Institute of Clinical Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Rongyao Zhang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Hailong Yuan
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Jianli Xu
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Kaile Zhang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Songqing Qi
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Xue Zhang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Ming Jiang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
40
|
Fang W, Wang J, Ma X, Shao N, Ye K, Zhang D, Shi C, Luo L. A Progressively Disassembled DNA Repair Inhibitors Nanosystem for the Treatment of BRCA Wild-Type Triple-Negative Breast Cancer. Int J Nanomedicine 2023; 18:6001-6019. [PMID: 37901361 PMCID: PMC10612513 DOI: 10.2147/ijn.s426639] [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: 07/10/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023] Open
Abstract
Background Olaparib, a poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor has demonstrated promising efficacy in patients with triple-negative breast cancer (TNBC) carrying breast cancer gene (BRCA) mutations. However, its impact on BRCA wild-type (BRCAwt) TNBC is limited. Hence, it is crucial to sensitize BRCAwt TNBC cells to olaparib for effective clinical practice. Novobiocin, a DNA polymerase theta (POLθ) inhibitor, exhibits sensitivity towards BRCA-mutated cancer cells that have acquired resistance to PARP inhibitors. Although both of these DNA repair inhibitors demonstrate therapeutic efficacy in BRCA-mutated cancers, their nanomedicine formulations' antitumor effects on wild-type cancer remain unclear. Furthermore, ensuring effective drug accumulation and release at the cancer site is essential for the clinical application of olaparib. Materials and Methods Herein, we designed a progressively disassembled nanosystem of DNA repair inhibitors as a novel strategy to enhance the effectiveness of olaparib in BRCAwt TNBC. The nanosystem enabled synergistic delivery of two DNA repair inhibitors olaparib and novobiocin, within an ultrathin silica framework interconnected by disulfide bonds. Results The designed nanosystem demonstrated remarkable capabilities, including long-term molecular storage and specific drug release triggered by the tumor microenvironment. Furthermore, the nanosystem exhibited potent inhibitory effects on cell viability, enhanced accumulation of DNA damage, and promotion of apoptosis in BRCAwt TNBC cells. Additionally, the nanosystem effectively accumulated within BRCAwt TNBC, leading to significant growth inhibition and displaying vascular regulatory abilities as assessed by magnetic resonance imaging (MRI). Conclusion Our results provided the inaugural evidence showcasing the potential of a progressively disassembled nanosystem of DNA repair inhibitors, as a promising strategy for the treatment of BRCA wild-type triple-negative breast cancer.
Collapse
Affiliation(s)
- Weimin Fang
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiaocong Ma
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Ni Shao
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Kunlin Ye
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Dong Zhang
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Changzheng Shi
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| | - Liangping Luo
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
41
|
Li C, Deng T, Cao J, Zhou Y, Luo X, Feng Y, Huang H, Liu J. Identifying ITGB2 as a Potential Prognostic Biomarker in Ovarian Cancer. Diagnostics (Basel) 2023; 13:diagnostics13061169. [PMID: 36980477 PMCID: PMC10047357 DOI: 10.3390/diagnostics13061169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Epithelial ovarian cancer is by far the most lethal gynecological malignancy. The exploration of promising immunomarkers to predict prognosis in ovarian cancer patients remains challenging. In our research, we carried out an integrated bioinformatic analysis of genome expressions and their immune characteristics in the ovarian cancer microenvironment with validation in different experiments. We filtrated 332 differentially expressed genes with 10 upregulated hub genes from the Gene Expression Omnibus database. These genes were closely related to ovarian tumorigenesis. Subsequently, the survival and immune infiltration analysis demonstrated that the upregulation of five candidate genes, ITGB2, VEGFA, CLDN4, OCLN, and SPP1, were correlated with an unfavorable clinical outcome and increased immune cell infiltration in ovarian cancer. Of these genes, ITGB2 tended to be the gene most correlated with various immune cell infiltrations and had a strong correlation with significant M2 macrophages infiltration (r = 0.707, p = 4.71 × 10-39), while it had a moderate correlation with CD4+/CD8+ T cells and B cells. This characteristic explains why the high expression of ITGB2 was accompanied by immune activation but did not reverse carcinogenesis. Additionally, we confirmed that ITGB2 was over-expressed in ovarian cancer tissues and was mainly located in cytoplasm, detected by Western blotting and the immunohistochemical method. In summary, ITGB2 may serve as a prognostic immunomarker for ovarian cancer patients.
Collapse
Affiliation(s)
- Chanyuan Li
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Ting Deng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Junya Cao
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yun Zhou
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiaolin Luo
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yanling Feng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - He Huang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Jihong Liu
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|