1
|
Wang R, Kumar P, Reda M, Wallstrum AG, Crumrine NA, Ngamcherdtrakul W, Yantasee W. Nanotechnology Applications in Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308639. [PMID: 38126905 PMCID: PMC11493329 DOI: 10.1002/smll.202308639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Next-generation cancer treatments are expected not only to target cancer cells but also to simultaneously train immune cells to combat cancer while modulating the immune-suppressive environment of tumors and hosts to ensure a robust and lasting response. Achieving this requires carriers that can codeliver multiple therapeutics to the right cancer and/or immune cells while ensuring patient safety. Nanotechnology holds great potential for addressing these challenges. This article highlights the recent advances in nanoimmunotherapeutic development, with a focus on breast cancer. While immune checkpoint inhibitors (ICIs) have achieved remarkable success and lead to cures in some cancers, their response rate in breast cancer is low. The poor response rate in solid tumors is often associated with the low infiltration of anti-cancer T cells and an immunosuppressive tumor microenvironment (TME). To enhance anti-cancer T-cell responses, nanoparticles are employed to deliver ICIs, bispecific antibodies, cytokines, and agents that induce immunogenic cancer cell death (ICD). Additionally, nanoparticles are used to manipulate various components of the TME, such as immunosuppressive myeloid cells, macrophages, dendritic cells, and fibroblasts to improve T-cell activities. Finally, this article discusses the outlook, challenges, and future directions of nanoimmunotherapeutics.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Pramod Kumar
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Noah A. Crumrine
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| |
Collapse
|
2
|
Zhou Y, Na C, Li Z. Novel insights into immune cells modulation of tumor resistance. Crit Rev Oncol Hematol 2024; 202:104457. [PMID: 39038527 DOI: 10.1016/j.critrevonc.2024.104457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024] Open
Abstract
Tumor resistance poses a significant challenge to effective cancer treatment, making it imperative to explore new therapeutic strategies. Recent studies have highlighted the profound involvement of immune cells in the development of tumor resistance. Within the tumor microenvironment, macrophages undergo polarization into the M2 phenotype, thus promoting the emergence of drug-resistant tumors. Neutrophils contribute to tumor resistance by forming extracellular traps. While T cells and natural killer (NK) cells exert their impact through direct cytotoxicity against tumor cells. Additionally, dendritic cells (DCs) have been implicated in preventing tumor drug resistance by stimulating T cell activation. In this review, we provide a comprehensive summary of the current knowledge regarding immune cell-mediated modulation of tumor resistance at the molecular level, with a particular focus on macrophages, neutrophils, DCs, T cells, and NK cells. The targeting of immune cell modulation exhibits considerable potential for addressing drug resistance, and an in-depth understanding of the molecular interactions between immune cells and tumor cells holds promise for the development of innovative therapies. Furthermore, we explore the clinical implications of these immune cells in the treatment of drug-resistant tumors. This review emphasizes the exploration of novel approaches that harness the functional capabilities of immune cells to effectively overcome drug-resistant tumors.
Collapse
Affiliation(s)
- Yi Zhou
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Chuhan Na
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhigang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen 518107, China.
| |
Collapse
|
3
|
Dang BTN, Duwa R, Lee S, Kwon TK, Chang JH, Jeong JH, Yook S. Targeting tumor-associated macrophages with mannosylated nanotherapeutics delivering TLR7/8 agonist enhances cancer immunotherapy. J Control Release 2024; 372:587-608. [PMID: 38942083 DOI: 10.1016/j.jconrel.2024.06.062] [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: 02/20/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Tumor-associated macrophages (TAMs) constitute 50-80% of stromal cells in most solid tumors with high mortality and poor prognosis. Tumor-infiltrating dendritic cells (TIDCs) and TAMs are key components mediating immune responses within the tumor microenvironment (TME). Considering their refractory properties, simultaneous remodeling of TAMs and TIDCs is a potential strategy of boosting tumor immunity and restoring immunosurveillance. In this study, mannose-decorated poly(lactic-co-glycolic acid) nanoparticles loading with R848 (Man-pD-PLGA-NP@R848) were prepared to dually target TAMs and TIDCs for efficient tumor immunotherapy. The three-dimensional (3D) cell culture model can simulate tumor growth as influenced by the TME and its 3D structural arrangement. Consequently, cancer spheroids enriched with tumor-associated macrophages (TAMs) were fabricated to assess the therapeutic effectiveness of Man-pD-PLGA-NP@R848. In the TME, Man-pD-PLGA-NP@R848 targeted both TAMs and TIDCs in a mannose receptor-mediated manner. Subsequently, Man-pD-PLGA-NP@R848 released R848 to activate Toll-like receptors 7 and 8, following dual-reprograming of TIDCs and TAMs. Man-pD-PLGA-NP@R848 could uniquely reprogram TAMs into antitumoral phenotypes, decrease angiogenesis, reprogram the immunosuppressive TME from "cold tumor" into "hot tumor", with high CD4+ and CD8+ T cell infiltration, and consequently hinder tumor development in B16F10 tumor-bearing mice. Therefore, dual-reprograming of TIDCs and TAMs with the Man-pD-PLGA-NP@R848 is a promising cancer immunotherapy strategy.
Collapse
Affiliation(s)
- Bao-Toan Nguyen Dang
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ramesh Duwa
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Jae-Hoon Chang
- College of Pharmacy, Yeungnam University, Gyeongbuk 38541, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
4
|
Turkmen Koc SN, Rezaei Benam S, Aral IP, Shahbazi R, Ulubayram K. Gold nanoparticles-mediated photothermal and photodynamic therapies for cancer. Int J Pharm 2024; 655:124057. [PMID: 38552752 DOI: 10.1016/j.ijpharm.2024.124057] [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: 11/17/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Cancer remains one of the major causes of death globally, with one out of every six deaths attributed to the disease. The impact of cancer is felt on psychological, physical, and financial levels, affecting individuals, communities, and healthcare institutions. Conventional cancer treatments have many challenges and inadequacies. Nanomedicine, however, presents a promising solution by not only overcoming these problems but also offering the advantage of combined therapy for treatment-resistant cancers. Nanoparticles specifically engineered for use in nanomedicine can be efficiently targeted to cancer cells through a combination of active and passive techniques, leading to superior tumor-specific accumulation, enhanced drug availability, and reduced systemic toxicity. Among various nanoparticle formulations designed for cancer treatment, gold nanoparticles have gained prominence in the field of nanomedicine due to their photothermal, photodynamic, and immunologic effects without the need for photosensitizers or immunotherapeutic agents. To date, there is no comprehensive literature review that focuses on the photothermal, photodynamic, and immunologic effects of gold nanoparticles. In this review, significant attention has been devoted to examining the parameters pertaining to the structure of gold nanoparticles and laser characteristics, which play a crucial role in influencing the efficacy of photothermal therapy (PTT) and photodynamic therapy (PDT). Moreover, this article provides insights into the success of PTT and PDT mediated by gold nanoparticles in primary cancer treatment, as well as the immunological effects of PTT and PDT on metastasis and recurrence, providing a promising strategy for cancer therapy. In summary, gold nanoparticles, with their unique properties, have the potential for clinical application in various cancer therapies, including the treatment of primary cancer, recurrence and metastasis.
Collapse
Affiliation(s)
- Seyma Nur Turkmen Koc
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye
| | - Sanam Rezaei Benam
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Ipek Pınar Aral
- Department of Radiation Oncology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara Bilkent City Hospital, Ankara, Türkiye
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA; Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, USA; Brown Center for Immunotherapy, Indiana University School of Medicine, Indianapolis, USA.
| | - Kezban Ulubayram
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye; Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye; Department of Bioengineering, Hacettepe University, Ankara, Türkiye.
| |
Collapse
|
5
|
Fan Z, Cui Y, Chen L, Liu P, Duan W. 23-Hydroxybetulinic acid attenuates 5-fluorouracil resistance of colorectal cancer by modulating M2 macrophage polarization via STAT6 signaling. Cancer Immunol Immunother 2024; 73:83. [PMID: 38554148 PMCID: PMC10981607 DOI: 10.1007/s00262-024-03662-0] [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: 01/01/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
Macrophage polarization is closely associated with the inflammatory processes involved in the development and chemoresistance of colorectal cancer (CRC). M2 macrophages, the predominant subtype of tumor-associated macrophages (TAMs) in a wide variety of malignancies, have been demonstrated to promote the resistance of CRC to multiple chemotherapeutic drugs, such as 5-fluorouracil (5-FU). In our study, we investigated the potential of 23-Hydroxybetulinic Acid (23-HBA), a significant active component of Pulsatilla chinensis (P. chinensis), to inhibit the polarization of M2 macrophages induced by IL-4. Our results showed that 23-HBA reduced the expression of M2 specific marker CD206, while downregulating the mRNA levels of M2 related genes (CD206, Arg1, IL-10, and CCL2). Additionally, 23-HBA effectively attenuated the inhibitory effects of the conditioned medium from M2 macrophages on apoptosis in colorectal cancer SW480 cells. Mechanistically, 23-HBA prevented the phosphorylation and nuclear translocation of the STAT6 protein, resulting in the inhibition of IL-10 release in M2 macrophages. Moreover, it interfered with the activation of the IL-10/STAT3/Bcl-2 signaling pathway in SW480 cells, ultimately reducing M2 macrophage-induced resistance to 5-FU. Importantly, depleting STAT6 expression in macrophages abolished the suppressive effect of 23-HBA on M2 macrophage polarization, while also eliminating its ability to decrease M2 macrophage-induced 5-FU resistance in cancer cells. Furthermore, 23-HBA significantly diminished the proportion of M2 macrophages in the tumor tissues of colorectal cancer mice, simultaneously enhancing the anti-cancer efficacy of 5-FU. The findings presented in this study highlight the capacity of 23-HBA to inhibit M2 macrophage polarization, a process that contributes to reduced 5-FU resistance in colorectal cancer.
Collapse
Affiliation(s)
- Zeping Fan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Yaru Cui
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, Jiangxi, China
| | - Lanying Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China.
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China.
- Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, Jiangxi, China.
| | - Peng Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Wenbin Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
6
|
Zhang J, Dong Y, Di S, Xie S, Fan B, Gong T. Tumor associated macrophages in esophageal squamous carcinoma: Promising therapeutic implications. Biomed Pharmacother 2023; 167:115610. [PMID: 37783153 DOI: 10.1016/j.biopha.2023.115610] [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: 08/16/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023] Open
Abstract
Esophageal squamous carcinoma (ESCC) is a prevalent and highly lethal malignant tumor, with a five-year survival rate of approximately 20 %. Tumor-associated macrophages (TAMs) are the most prominent immune cells in the tumor microenvironment (TME), comprising over 50 % of the tumor volume. TAMs can be polarized into two distinct phenotypes, M1-type and M2-type, through interactions with cancer cells. M2-type TAMs are more abundant than M1-type TAMs in the TME, contributing to tumor progression, such as tumor cell survival and the construction of an immunosuppressive environment. This review focuses on the role of TAMs in ESCC, including their polarization, impact on tumor proliferation, angiogenesis, invasion, migration, therapy resistance, and immunosuppression. In addition, we discuss the potential of targeting TAMs for clinical therapy in ESCC. A thorough comprehension of the molecular biology about TAMs is essential for the development of innovative therapeutic strategies to treat ESCC.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China; Department of Thoracic Surgery, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yanxin Dong
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China; Department of Thoracic Surgery, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shouyin Di
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Shun Xie
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Boshi Fan
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China.
| | - Taiqian Gong
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
7
|
Xu X, Chen J, Li W, Feng C, Liu Q, Gao W, He M. Immunology and immunotherapy in gastric cancer. Clin Exp Med 2023; 23:3189-3204. [PMID: 37322134 DOI: 10.1007/s10238-023-01104-2] [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: 01/16/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023]
Abstract
Gastric cancer is the fifth leading cause of cancer-related deaths worldwide. As the diagnosis of early gastric cancer is difficult, most patients are at a late stage of cancer progression when diagnosed. The current therapeutic approaches based on surgical or endoscopic resection and chemotherapy indeed improve patients' outcomes. Immunotherapy based on immune checkpoint inhibitors has opened a new era for cancer treatment, and the immune system of the host is reshaped to combat tumor cells and the strategy differs according to the patient's immune system. Thus, an in-depth understanding of the roles of various immune cells in the progression of gastric cancer is beneficial to application for immunotherapy and the discovery of new therapeutic targets. This review describes the functions of different immune cells in gastric cancer development, mainly focusing on T cells, B cells, macrophages, natural killer cells, dendritic cells, neutrophils as well as chemokines or cytokines secreted by tumor cells. And this review also discusses the latest advances in immune-related therapeutic approaches such as immune checkpoint inhibitors, CAR-T or vaccine, to reveal potential and promising strategies for gastric cancer treatment.
Collapse
Affiliation(s)
- Xiaqing Xu
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, People's Republic of China
| | - Jiaxing Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Wenxing Li
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, People's Republic of China
| | - Chenlu Feng
- Department of Cancer Center, Nanyang First People's Hospital, Nanyang, 473000, Henan, People's Republic of China
| | - Qian Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Wenfang Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Meng He
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, People's Republic of China.
| |
Collapse
|
8
|
Gan H, Huang X, Luo X, Li J, Mo B, Cheng L, Shu Q, Du Z, Tang H, Sun W, Wang L, Luo S, Yu S. A Mitochondria-Targeted Ferroptosis Inducer Activated by Glutathione-Responsive Imaging and Depletion for Triple Negative Breast Cancer Theranostics. Adv Healthc Mater 2023; 12:e2300220. [PMID: 37204240 DOI: 10.1002/adhm.202300220] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/19/2023] [Indexed: 05/20/2023]
Abstract
Ferroptosis is a new type of iron-dependent programmed cell death characterized by glutathione (GSH) depletion, selenoprotein glutathione peroxidase 4 (GPX4) inactivation, and lipid peroxides accumulation. Mitochondria, as the main source of intracellular energy supply and reactive oxygen species (ROS) generation, play a central role in oxidative phosphorylation and redox homeostasis. Therefore, targeting cancer-cell mitochondria and attacking redox homeostasis is expected to induce robust ferroptosis-mediated anticancer effects. In this work, a theranostic ferroptosis inducer (IR780-SPhF), which can simultaneously achieve the imaging and therapy of triple-negative breast cancer (TNBC) by targeting mitochondria is presented. It is developed from a mitochondria-targeting small molecule (IR780) with cancer-preferential accumulation, enabling it to react with GSH by nucleophilic substitution, resulting in mitochondrial GSH depletion and redox imbalance. More interestingly, IR780-SPhF exhibits GSH-responsive near-infrared fluorescence emission and photoacoustic imaging characteristics, further facilitating diagnosis and treatment with real-time monitoring of TNBC with a highly elevated GSH level. Both in vitro and in vivo results demonstrate that IR780-SPhF exhibits potent anticancer effect, which is significantly stronger than cyclophosphamide, a classic drug commonly recommended for TNBC patients in clinic. Hence, the reported mitochondria-targeted ferroptosis inducer may represent a promising candidate and a prospective strategy for efficient cancer treatment.
Collapse
Affiliation(s)
- Hongbo Gan
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Xie Huang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xi Luo
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Jinlin Li
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Banghui Mo
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Lizhi Cheng
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Qiuxia Shu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Zaizhi Du
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hong Tang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Chongqing Key Laboratory of Cytomics, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Chongqing Key Laboratory of Cytomics, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Shenglin Luo
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Songtao Yu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| |
Collapse
|
9
|
Lim GH, An JH, Park SM, Youn GH, Oh YI, Seo KW, Youn HY. Macrophage induces anti-cancer drug resistance in canine mammary gland tumor spheroid. Sci Rep 2023; 13:10394. [PMID: 37369757 DOI: 10.1038/s41598-023-37311-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023] Open
Abstract
Tumor-associated macrophages (TAMs) play an important role in the tumor microenvironment by producing cytokines and growth factors. Furthermore, TAMs play multifunctional roles in tumor progression, immune regulation, metastasis, angiogenesis, and chemoresistance. Hypoxia in the tumor microenvironment induces tumor-supporting transformation of TAMs, which enhances tumor malignancy through developing anti-cancer resistance, for example. In this study, a hybrid spheroid model of canine mammary gland tumor (MGT) cell lines (CIPp and CIPm) and canine macrophages (DH82) was established. The effects of hypoxia induced by the spheroid culture system on the anti-cancer drug resistance of canine MGT cells were investigated. A hybrid spheroid was created using an ultralow-adhesion plate. The interactions between canine MGT cells and DH82 were investigated using a co-culture method. When co-cultured with DH82, cell viability and expression levels of tumor growth factors and multi-drug resistance genes were increased in canine MGT cells under doxorubicin. Additionally, doxorubicin-induced apoptosis and G2/M cell cycle arrest were attenuated in canine MGT cells co-cultured with DH82. In conclusion, the hybrid spheroid model established in this study reflects the hypoxic TME, allowing DH82 to induce anti-cancer drug resistance in canine MGT cells.
Collapse
Affiliation(s)
- Ga-Hyun Lim
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ju-Hyun An
- Department of Veterinary Emergency and Critical Care Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Su-Min Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ga-Hee Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ye-In Oh
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoung-Won Seo
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
10
|
Kesharwani P, Ma R, Sang L, Fatima M, Sheikh A, Abourehab MAS, Gupta N, Chen ZS, Zhou Y. Gold nanoparticles and gold nanorods in the landscape of cancer therapy. Mol Cancer 2023; 22:98. [PMID: 37344887 DOI: 10.1186/s12943-023-01798-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
Cancer is a grievous disease whose treatment requires a more efficient, non-invasive therapy, associated with minimal side effects. Gold nanoparticles possessing greatly impressive optical properties have been a forerunner in bioengineered cancer therapy. This theranostic system has gained immense popularity and finds its application in the field of molecular detection, biological imaging, cancer cell targeting, etc. The photothermal property of nanoparticles, especially of gold nanorods, causes absorption of the light incident by the light source, and transforms it into heat, resulting in tumor cell destruction. This review describes the different optical features of gold nanoparticles and summarizes the advance research done for the application of gold nanoparticles and precisely gold nanorods for combating various cancers including breast, lung, colon, oral, prostate, and pancreatic cancer.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, China
| | - Mahak Fatima
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Neelima Gupta
- Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York City, NY, 11439, USA
| | - Yun Zhou
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
12
|
Cui Y, Xu Y, Li Y, Sun Y, Hu J, Jia J, Li X. Antibody Drug Conjugates of Near-Infrared Photoimmunotherapy (NIR-PIT) in Breast Cancers. Technol Cancer Res Treat 2023; 22:15330338221145992. [PMID: 36734039 PMCID: PMC9903039 DOI: 10.1177/15330338221145992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Worldwide, the incidence rate of breast cancer is the highest in women. Approximately 2.3 million people were newly diagnosed and 0.685 million were dead of breast cancer in 2020, which continues to grow. Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with a higher risk of recurrence and metastasis, but disappointly, there are no effective and specific therapies clinically, especially for patients presenting with metastatic diseases. Therefore, it is urgent to develop a new type of cancer therapy for survival improvisation and adverse effects alleviation of breast cancers. Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed, photochemistry-based cancer therapy. It was drive by an antibody-photoabsorber conjugate (APC) which is triggered by near-infrared light. The key part of APC is a cancer-targeting monoclonal antibody (mAb) that can bind to receptors or antigens on the surface of tumor cells. Because of this targeted conjugate accumulation, subsequent deployment of focal NIR-light results in functional damage on the targeted cell membranes without harming the immediately adjacent receptor-negative cells and evokes a kind of photochemical, speedy, and highly specific immunogenic cell death (ICD) of cancer cells with corresponding antigens. Subsequently, immature dendritic cells adjacent to dying cancer cells will become mature, further inducing a host-oriented anti-cancer immune response, complicatedly and comprehensively. Currently, NIR-PIT has progressed into phase 3 clinical trial for recurrent head and neck cancer. And preclinical studies have illustrated strong therapeutic efficacy of NIR-PIT targeting various molecular receptors overexpressed in breast cancer cells, including EGFR, HER2, CD44c, CD206, ICAM-1 and FAP-α. Thereby, NIR-PIT is in early trials, but appears to be a promising breast cancer therapy and moving into the future. Here, we present the specific advantages and discuss the most recent preclinical studies against several transmembrane proteins of NIR-PIT in breast cancers.
Collapse
Affiliation(s)
- Yingshu Cui
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China
| | - Yuanyuan Xu
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Laser, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yi Li
- Medical School of Chinese PLA, Beijing, China,Department of Laser, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuanyuan Sun
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jia Hu
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jia Jia
- Department of Oncology, the Seventh Medical Center, Chinese PLA General Hospital, Beijing, China,Jia Jia, Department of Oncology, the Seventh Medical Centre, Chinese PLA General Hospital, Beijing 100700, China.
| | - Xiaosong Li
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China,Xiaosong Li, Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China.
| |
Collapse
|
13
|
Barnestein R, Galland L, Kalfeist L, Ghiringhelli F, Ladoire S, Limagne E. Immunosuppressive tumor microenvironment modulation by chemotherapies and targeted therapies to enhance immunotherapy effectiveness. Oncoimmunology 2022; 11:2120676. [PMID: 36117524 PMCID: PMC9481153 DOI: 10.1080/2162402x.2022.2120676] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
With the rapid clinical development of immune checkpoint inhibitors (ICIs), the standard of care in cancer management has evolved rapidly. However, immunotherapy is not currently beneficial for all patients. In addition to intrinsic tumor factors, other etiologies of resistance to ICIs arise from the complex interplay between cancer and its microenvironment. Recognition of the essential role of the tumor microenvironment (TME) in cancer progression has led to a shift from a tumor-cell-centered view of cancer development, to the concept of a complex tumor ecosystem that supports tumor growth and metastatic dissemination. The expansion of immunosuppressive cells represents a cardinal strategy deployed by tumor cells to escape detection and elimination by the immune system. Regulatory T lymphocytes (Treg), myeloid-derived suppressor cells (MDSCs), and type-2 tumor-associated macrophages (TAM2) are major components of these inhibitory cellular networks, with the ability to suppress innate and adaptive anticancer immunity. They therefore represent major impediments to anticancer therapies, particularly immune-based interventions. Recent work has provided evidence that, beyond their direct cytotoxic effects on cancer cells, several conventional chemotherapeutic (CT) drugs and agents used in targeted therapies (TT) can promote the elimination or inactivation of suppressive immune cells, resulting in enhanced antitumor immunity. In this review, we will analyze findings pertaining to this concept, discuss the possible molecular bases underlying the selective targeting of these immunosuppressive cells by antineoplastic agents (CT and/or TT), and consider current challenges and future prospects related to the integration of these molecules into more efficient anticancer strategies, in the era of immunotherapy.
Collapse
Affiliation(s)
- Robby Barnestein
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
| | - Loïck Galland
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
| | - Laura Kalfeist
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - François Ghiringhelli
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Sylvain Ladoire
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Emeric Limagne
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| |
Collapse
|
14
|
Potential Nanotechnology-Based Therapeutics to Prevent Cancer Progression through TME Cell-Driven Populations. Pharmaceutics 2022; 15:pharmaceutics15010112. [PMID: 36678741 PMCID: PMC9864587 DOI: 10.3390/pharmaceutics15010112] [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: 11/11/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a high risk of metastasis and therapeutic resistance. These issues are closely linked to the tumour microenvironment (TME) surrounding the tumour tissue. The association between residing TME components with tumour progression, survival, and metastasis has been well elucidated. Focusing on cancer cells alone is no longer considered a viable approach to therapy; thus, there is a high demand for TME targeting. The benefit of using nanoparticles is their preferential tumour accumulation and their ability to target TME components. Several nano-based platforms have been investigated to mitigate microenvironment-induced angiogenesis, therapeutic resistance, and tumour progression. These have been achieved by targeting mesenchymal originating cells (e.g., cancer-associated fibroblasts, adipocytes, and stem cells), haematological cells (e.g., tumour-associated macrophages, dendritic cells, and myeloid-derived suppressor cells), and the extracellular matrix within the TME that displays functional and architectural support. This review highlights the importance of nanotechnology-based therapeutics as a promising approach to target the TME and improve treatment outcomes for TNBC patients, which can lead to enhanced survival and quality of life. The role of different nanotherapeutics has been explored in the established TME cell-driven populations.
Collapse
|
15
|
Bhandari M, Raj S, Kumar A, Kaur DP. Bibliometric analysis on exploitation of biogenic gold and silver nanoparticles in breast, ovarian and cervical cancer therapy. Front Pharmacol 2022; 13:1035769. [PMID: 36618941 PMCID: PMC9818348 DOI: 10.3389/fphar.2022.1035769] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Multifunctional nanoparticles are being formulated to overcome the side effects associated with anticancer drugs as well as conventional drug delivery systems. Cancer therapy has gained the advancement due to various pragmatic approaches with better treatment outcomes. The metal nanostructures such as gold and silver nanoparticles accessible via eco-friendly method provide amazing characteristics in the field of diagnosis and therapy towards cancer diseases. The environmental friendly approach has been proposed as a substitute to minimize the use of hazardous compounds associated in chemical synthesis of nanoparticles. In this attempt, researchers have used various microbes, and plant-based agents as reducing agents. In the last 2 decades various papers have been published emphasizing the benefits of the eco-friendly approach and advantages over the traditional method in the cancer therapy. Despite of various reports and published research papers, eco-based nanoparticles do not seem to find a way to clinical translation for cancer treatment. Present review enumerates the bibliometric data on biogenic silver and gold nanoparticles from Clarivate Analytics Web of Science (WoS) and Scopus for the duration 2010 to 2022 for cancer treatment with a special emphasis on breast, ovarian and cervical cancer. Furthermore, this review covers the recent advances in this area of research and also highlights the obstacles in the journey of biogenic nanodrug from clinic to market.
Collapse
Affiliation(s)
- Meena Bhandari
- Department of Chemistry, School of Basic and Applied Sciences, K.R Mangalam University, Gurugram, India
| | - Seema Raj
- Department of Chemistry, School of Basic and Applied Sciences, K.R Mangalam University, Gurugram, India,*Correspondence: Seema Raj, ,
| | - Ashwani Kumar
- Department of Computer Sciences, School of Engineering and Technology, K.R Mangalam University, Gurugram, India
| | - Dilraj Preet Kaur
- Department of Physics, School of Basic and Applied Sciences, K.R Mangalam University, Gurugram, India
| |
Collapse
|
16
|
Pei Z, Chen S, Ding L, Liu J, Cui X, Li F, Qiu F. Current perspectives and trend of nanomedicine in cancer: A review and bibliometric analysis. J Control Release 2022; 352:211-241. [PMID: 36270513 DOI: 10.1016/j.jconrel.2022.10.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022]
Abstract
The limitations of traditional cancer treatments are driving the creation and development of new nanomedicines. At present, with the rapid increase of research on nanomedicine in the field of cancer, there is a lack of intuitive analysis of the development trend, main authors and research hotspots of nanomedicine in the field of cancer, as well as detailed elaboration of possible research hotspots. In this review, data collected from the Web of Science Core Collection database between January 1st, 2000, and December 31st, 2021, were subjected to a bibliometric analysis. The co-authorship, co-citation, and co-occurrence of countries, institutions, authors, literature, and keywords in this subject were examined using VOSviewer, Citespace, and a well-known online bibliometrics platform. We collected 19,654 published papers, China produced the most publications (36.654%, 7204), followed by the United States (29.594%, 5777), and India (7.780%, 1529). An interesting fact is that, despite China having more publications than the United States, the United States still dominates this field, having the highest H-index and the most citations. Acs Nano, Nano Letters, and Biomaterials are the top three academic publications that publish articles on nanomedicine for cancer out of a total of 7580 academic journals. The most significant increases were shown for the keywords "cancer nanomedicine", "tumor microenvironment", "nanoparticles", "prodrug", "targeted nanomedicine", "combination", and "cancer immunotherapy" indicating the promising area of research. Meanwhile, the development prospects and challenges of nanomedicine in cancer are also discussed and provided some solutions to the major obstacles.
Collapse
Affiliation(s)
- Zerong Pei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuting Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Liqin Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xinyi Cui
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
17
|
Xia Y, Yang R, Zhu J, Wang H, Li Y, Fan J, Fu C. Engineered nanomaterials trigger abscopal effect in immunotherapy of metastatic cancers. Front Bioeng Biotechnol 2022; 10:890257. [PMID: 36394039 PMCID: PMC9643844 DOI: 10.3389/fbioe.2022.890257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 10/14/2022] [Indexed: 11/24/2022] Open
Abstract
Despite advances in cancer treatment, metastatic cancer is still the main cause of death in cancer patients. At present, the treatment of metastatic cancer is limited to palliative care. The abscopal effect is a rare phenomenon in which shrinkage of metastatic tumors occurs simultaneously with the shrinkage of a tumor receiving localized treatment, such as local radiotherapy or immunotherapy. Immunotherapy shows promise for cancer treatment, but it also leads to consequences such as low responsiveness and immune-related adverse events. As a promising target-based approach, intravenous or intratumoral injection of nanomaterials provides new opportunities for improving cancer immunotherapy. Chemically modified nanomaterials may be able to trigger the abscopal effect by regulating immune cells. This review discusses the use of nanomaterials in killing metastatic tumor cells through the regulation of immune cells and the prospects of such nanomaterials for clinical use.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ruohan Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuehong Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Changfeng Fu,
| |
Collapse
|
18
|
Cınar V, Hamurcu Z, Guler A, Nurdinov N, Ozpolat B. Serotonin 5-HT7 receptor is a biomarker poor prognostic factor and induces proliferation of triple-negative breast cancer cells through FOXM1. Breast Cancer 2022; 29:1106-1120. [PMID: 36006564 DOI: 10.1007/s12282-022-01391-9] [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: 04/14/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer and associated with poor prognosis and shorter survival due to significant genetic heterogeneity, drug resistance and lack of effective targeted therapeutics. Therefore, novel molecular targets and therapeutic strategies are needed to improve patient survival. Serotonin (5-hydroxytryptamine, 5-HT) has been shown to induce growth stimulatory effects in breast cancer. However, the molecular mechanisms by which 5-HT exerts its oncogenic effects in TNBC still are not well understood. METHODS Normal breast epithelium (MCF10A) and two TNBC cells (MDA-MB-231, BT-546) and MCF-7 cells (ER +) were used to investigate effects of 5-HT7 receptor. Small interfering RNA (siRNA)-based knockdown and metergoline (5-HT7 antagonist) were used to inhibit the activity of 5-HT7. Cell proliferation and colony formation were evaluated using MTS cell viability and colony formation assays, respectively. Western blotting was used to investigate 5-HT7, FOXM1 and its downstream targets protein expressions. RESULTS We demonstrated that 5-HT induces cell proliferation of TNBC cells and expression of 5-HT7 receptor and FOXM1 oncogenic transcription factor. We found that expression of 5-HT7 receptor is up-regulated in TNBC cells and higher 5-HT7 receptor expression is associated with poor patient prognosis and shorter patient survival. Genetic and pharmacological inhibition of 5-HT7 receptor by siRNA and metergoline, respectively, suppressed TNBC cell proliferation and FOXM1 and its downstream mediators, including eEF2-Kinase (eEF2K) and cyclin-D1. CONCLUSION Our findings suggest for the first time that the 5-HT7 receptor promotes FOXM1, eEF2K and cyclin D1 signaling to support TNBC cell proliferation; thus, inhibition of 5-HT7 receptor/FOXM1 signaling may be used as a potential therapeutic strategy for targeting TNBC. 5-HT induces cell proliferation of TNBC cells through 5-HT7 receptor signaling. Also, genetic and pharmacological inhibition of 5-HT7 by RNAi (siRNA) and metergoline HTR7 antagonist, respectively inhibits FOXM1 oncogenic transcription factor and suppresses TNBC cell proliferation.
Collapse
Affiliation(s)
- Venhar Cınar
- Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey.,Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Zuhal Hamurcu
- Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey.,Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 422, Houston, TX, 77030, USA
| | - Ahsen Guler
- Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey.,Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Nursultan Nurdinov
- Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey.,Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 422, Houston, TX, 77030, USA. .,RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Houston Methodist Neal Cancer, Houston, TX, USA.
| |
Collapse
|
19
|
Guha L, Bhat IA, Bashir A, Rahman JU, Pottoo FH. Nanotechnological Approaches for the Treatment of Triple-Negative Breast Cancer: A Comprehensive Review. Curr Drug Metab 2022; 23:781-799. [PMID: 35676850 DOI: 10.2174/1389200223666220608144551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 03/10/2022] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most prevalent cancer in women around the world, having a sudden spread nowadays because of the poor sedentary lifestyle of people. Comprising several subtypes, one of the most dangerous and aggressive ones is triple-negative breast cancer or TNBC. Even though conventional surgical approaches like single and double mastectomy and preventive chemotherapeutic approaches are available, they are not selective to cancer cells and are only for symptomatic treatment. A new branch called nanotechnology has emerged in the last few decades that offers various novel characteristics, such as size in nanometric scale, enhanced adherence to multiple targeting moieties, active and passive targeting, controlled release, and site-specific targeting. Among various nanotherapeutic approaches like dendrimers, lipid-structured nanocarriers, carbon nanotubes, etc., nanoparticle targeted therapeutics can be termed the best among all for their specific cytotoxicity to cancer cells and increased bioavailability to a target site. This review focuses on the types and molecular pathways involving TNBC, existing treatment strategies, various nanotechnological approaches like exosomes, carbon nanotubes, dendrimers, lipid, and carbon-based nanocarriers, and especially various nanoparticles (NPs) like polymeric, photodynamic, peptide conjugated, antibody-conjugated, metallic, inorganic, natural product capped, and CRISPR based nanoparticles already approved for treatment or are under clinical and pre-clinical trials for TNBC.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Mohali, S.A.S Nagar, Punjab 160062, India
| | - Ishfaq Ahmad Bhat
- Northern Railway Hospital, Sri Mata Vaishno Devi, Katra, Reasi 182320, India
| | - Aasiya Bashir
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, J&K, India
| | - Jawad Ur Rahman
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
20
|
Schirrmann R, Erkelenz M, Lamers K, Sritharan O, Nachev M, Sures B, Schlücker S, Brandau S. Gold Nanorods Induce Endoplasmic Reticulum Stress and Autocrine Inflammatory Activation in Human Neutrophils. ACS NANO 2022; 16:11011-11026. [PMID: 35737452 DOI: 10.1021/acsnano.2c03586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Gold nanorods (AuNRs) are promising agents for diverse biomedical applications such as drug and gene delivery, bioimaging, and cancer treatment. Upon in vivo application, AuNRs quickly interact with cells of the immune system. On the basis of their strong intrinsic phagocytic activity, polymorphonuclear neutrophils (PMNs) are specifically equipped for the uptake of particulate materials such as AuNRs. Therefore, understanding the interaction of AuNRs with PMNs is key for the development of safe and efficient therapeutic applications. In this study, we investigated the uptake, intracellular processing, and cell biological response induced by AuNRs in PMNs. We show that uptake of AuNRs mainly occurs via phagocytosis and macropinocytosis with rapid deposition of AuNRs in endosomes within 5 min. Within 60 min, AuNR uptake induced an unfolded protein response (UPR) along with induction of inositol-requiring enzyme 1 α (IREα) and features of endoplasmic reticulum (ER) stress. This early response was followed by a pro-inflammatory autocrine activation loop that involves LOX1 upregulation on the cell surface and increased secretion of IL8 and MMP9. Our study provides comprehensive mechanistic insight into the interaction of AuNRs with immune cells and suggests potential targets to limit the unwanted immunopathological activation of PMNs during application of AuNRs.
Collapse
Affiliation(s)
- Ronja Schirrmann
- Department of Otorhinolaryngology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Michael Erkelenz
- Department of Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
| | - Kim Lamers
- Department of Otorhinolaryngology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Oliver Sritharan
- Department of Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
| | - Milen Nachev
- Department of Aquatic Ecology and Centre for Water and Environmental Research (ZWU), University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
| | - Bernd Sures
- Department of Aquatic Ecology and Centre for Water and Environmental Research (ZWU), University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
| | - Sebastian Schlücker
- Department of Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
- Center of Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
- Center of Medical Biotechnology (ZMB), University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
- University of Duisburg-Essen, Universitätsstraße 5, 451471 Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
- Center of Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany
- Center of Medical Biotechnology (ZMB), University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
- University of Duisburg-Essen, Universitätsstraße 5, 451471 Essen, Germany
- German Cancer Consortium, Partner Site Essen-Düsseldorf, 45147 Essen, Germany
| |
Collapse
|
21
|
A Sequential Stimulus-Responsive Nanoparticle Platform for Enhanced Tumor Growth Inhibition and Efficient Drug Delivery. J Pharm Innov 2022. [DOI: 10.1007/s12247-022-09653-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Shokooh MK, Emami F, Duwa R, Jeong JH, Yook S. Triple-negative breast cancer treatment meets nanoparticles: Current status and future direction. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
23
|
Xiao M, He J, Yin L, Chen X, Zu X, Shen Y. Tumor-Associated Macrophages: Critical Players in Drug Resistance of Breast Cancer. Front Immunol 2022; 12:799428. [PMID: 34992609 PMCID: PMC8724912 DOI: 10.3389/fimmu.2021.799428] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Drug resistance is one of the most critical challenges in breast cancer (BC) treatment. The occurrence and development of drug resistance are closely related to the tumor immune microenvironment (TIME). Tumor-associated macrophages (TAMs), the most important immune cells in TIME, are essential for drug resistance in BC treatment. In this article, we summarize the effects of TAMs on the resistance of various drugs in endocrine therapy, chemotherapy, targeted therapy, and immunotherapy, and their underlying mechanisms. Based on the current overview of the key role of TAMs in drug resistance, we discuss the potential possibility for targeting TAMs to reduce drug resistance in BC treatment, By inhibiting the recruitment of TAMs, depleting the number of TAMs, regulating the polarization of TAMs and enhancing the phagocytosis of TAMs. Evidences in our review support it is important to develop novel therapeutic strategies to target TAMs in BC to overcome the treatment of resistance.
Collapse
Affiliation(s)
- Maoyu Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguan Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
24
|
Emami F, Banstola A, Jeong JH, Yook S. Cetuximab-anchored gold nanorod mediated photothermal ablation of breast cancer cell in spheroid model embedded with tumor associated macrophage. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.10.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Dynamic increase of M2 macrophages is associated with disease progression of colorectal cancers following cetuximab-based treatment. Sci Rep 2022; 12:1678. [PMID: 35102212 PMCID: PMC8803829 DOI: 10.1038/s41598-022-05694-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
We aimed to investigate the dynamic changes of gene expression profiles and immune microenvironment linked to resistance to cetuximab-based treatments in patients with metastatic colorectal cancer (mCRC). A total of 106 patients with RAS-wild type mCRC who were treated with cetuximab-based treatments were included as the study population. RNA-sequencing and multiplexed immunohistochemistry were performed using paired or unpaired pre-treatment and post-treatment tumor tissues. Differentially expressed gene analysis of paired pre-treatment and post-treatment tumor tissues that develop acquired resistance (AR) identified the AR signature. Gene ontology analysis of the AR signature indicated enrichment of immune-related pathway genes. Among the immune subsets whose abundance was estimated by CIBERSORT, M2 macrophages showed the most prominent positive correlation with the expression of the AR signature. Among the post-treatment samples, progressive disease (PD) tumors showed a significantly higher abundance of M2 macrophages compared to non-PD tumors. These findings were validated by multiplexed immunohistochemistry analysis: the density of CD68+CD206+ M2 macrophages significantly increased at the time of PD following cetuximab-based treatment, whereas it did not consistently change in the tumor pairs of non-PD. In conclusion, a dynamic increase of M2 macrophages is associated with disease progression during cetuximab-based treatment of mCRCs. Targeting M2 macrophages is a promising immunotherapeutic strategy in this clinical context.
Collapse
|
26
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
27
|
Larionova I, Kazakova E, Gerashchenko T, Kzhyshkowska J. New Angiogenic Regulators Produced by TAMs: Perspective for Targeting Tumor Angiogenesis. Cancers (Basel) 2021; 13:cancers13133253. [PMID: 34209679 PMCID: PMC8268686 DOI: 10.3390/cancers13133253] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Since the targeting of a single pro-angiogenic factor fails to improve oncological disease outcome, significant efforts have been made to identify new pro-angiogenic factors that could compensate for the deficiency of current therapy or act independently as single drugs. Our review aims to present the state-of-the art for well-known and recently described factors produced by macrophages that induce and regulate angiogenesis. A number of positive and negative regulators of angiogenesis in the tumor microenvironment are produced by tumor-associated macrophages (TAMs). Accumulating evidence has indicated that, apart from the well-known angiogenic factors, there are plenty of novel angiogenesis-regulating proteins that belong to different classes. We summarize the data regarding the direct or indirect mechanisms of the interaction of these factors with endothelial cells during angiogenesis. We highlight the recent findings that explain the limitations in the efficiency of current anti-angiogenic therapy approaches. Abstract Angiogenesis is crucial to the supply of a growing tumor with nutrition and oxygen. Inhibition of angiogenesis is one of the main treatment strategies for colorectal, lung, breast, renal, and other solid cancers. However, currently applied drugs that target VEGF or receptor tyrosine kinases have limited efficiency, which raises a question concerning the mechanism of patient resistance to the already developed drugs. Tumor-associated macrophages (TAMs) were identified in the animal tumor models as a key inducer of the angiogenic switch. TAMs represent a potent source not only for VEGF, but also for a number of other pro-angiogenic factors. Our review provides information about the activity of secreted regulators of angiogenesis produced by TAMs. They include members of SEMA and S100A families, chitinase-like proteins, osteopontin, and SPARC. The COX-2, Tie2, and other factors that control the pro-angiogenic activity of TAMs are also discussed. We highlight how these recent findings explain the limitations in the efficiency of current anti-angiogenic therapy. Additionally, we describe genetic and posttranscriptional mechanisms that control the expression of factors regulating angiogenesis. Finally, we present prospects for the complex targeting of the pro-angiogenic activity of TAMs.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
- Correspondence: (I.L.); (J.K.)
| | - Elena Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
| | - Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 68167 Mannheim, Germany
- Correspondence: (I.L.); (J.K.)
| |
Collapse
|
28
|
Chojnacki K, Wińska P, Karatsai O, Koronkiewicz M, Milner-Krawczyk M, Wielechowska M, Rędowicz MJ, Bretner M, Borowiecki P. Synthesis of Novel Acyl Derivatives of 3-(4,5,6,7-Tetrabromo-1 H-benzimidazol-1-yl)propan-1-ols-Intracellular TBBi-Based CK2 Inhibitors with Proapoptotic Properties. Int J Mol Sci 2021; 22:6261. [PMID: 34200807 PMCID: PMC8230474 DOI: 10.3390/ijms22126261] [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: 05/13/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/09/2022] Open
Abstract
Protein kinase CK2 has been considered as an attractive drug target for anti-cancer therapy. The synthesis of N-hydroxypropyl TBBi and 2MeTBBi derivatives as well as their respective esters was carried out by using chemoenzymatic methods. Concomitantly with kinetic studies toward recombinant CK2, the influence of the obtained compounds on the viability of two human breast carcinoma cell lines (MCF-7 and MDA-MB-231) was evaluated using MTT assay. Additionally, an intracellular inhibition of CK2 as well as an induction of apoptosis in the examined cells after the treatment with the most active compounds were studied by Western blot analysis, phase-contrast microscopy and flow cytometry method. The results of the MTT test revealed potent cytotoxic activities for most of the newly synthesized compounds (EC50 4.90 to 32.77 µM), corresponding to their solubility in biological media. We concluded that derivatives with the methyl group decrease the viability of both cell lines more efficiently than their non-methylated analogs. Furthermore, inhibition of CK2 in breast cancer cells treated with the tested compounds at the concentrations equal to their EC50 values correlates well with their lipophilicity since derivatives with higher values of logP are more potent intracellular inhibitors of CK2 with better proapoptotic properties than their parental hydroxyl compounds.
Collapse
Affiliation(s)
- Konrad Chojnacki
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Patrycja Wińska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (O.K.); (M.J.R.)
| | - Mirosława Koronkiewicz
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Małgorzata Milner-Krawczyk
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Monika Wielechowska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (O.K.); (M.J.R.)
| | - Maria Bretner
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Paweł Borowiecki
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| |
Collapse
|
29
|
Cotechini T, Atallah A, Grossman A. Tissue-Resident and Recruited Macrophages in Primary Tumor and Metastatic Microenvironments: Potential Targets in Cancer Therapy. Cells 2021; 10:960. [PMID: 33924237 PMCID: PMC8074766 DOI: 10.3390/cells10040960] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages within solid tumors and metastatic sites are heterogenous populations with different developmental origins and substantially contribute to tumor progression. A number of tumor-promoting phenotypes associated with both tumor- and metastasis-associated macrophages are similar to innate programs of embryonic-derived tissue-resident macrophages. In contrast to recruited macrophages originating from marrow precursors, tissue-resident macrophages are seeded before birth and function to coordinate tissue remodeling and maintain tissue integrity and homeostasis. Both recruited and tissue-resident macrophage populations contribute to tumor growth and metastasis and are important mediators of resistance to chemotherapy, radiation therapy, and immune checkpoint blockade. Thus, targeting various macrophage populations and their tumor-promoting phenotypes holds therapeutic promise. Here, we discuss various macrophage populations as regulators of tumor progression, immunity, and immunotherapy. We provide an overview of macrophage targeting strategies, including therapeutics designed to induce macrophage depletion, impair recruitment, and induce repolarization. We also provide a perspective on the therapeutic potential for macrophage-specific acquisition of trained immunity as an anti-cancer agent and discuss the therapeutic potential of exploiting macrophages and their traits to reduce tumor burden.
Collapse
Affiliation(s)
- Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.A.); (A.G.)
| | | | | |
Collapse
|
30
|
Keihan Shokooh M, Emami F, Jeong JH, Yook S. Bio-Inspired and Smart Nanoparticles for Triple Negative Breast Cancer Microenvironment. Pharmaceutics 2021; 13:287. [PMID: 33671698 PMCID: PMC7926463 DOI: 10.3390/pharmaceutics13020287] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) with poor prognosis and aggressive nature accounts for 10-20% of all invasive breast cancer (BC) cases and is detected in as much as 15% of individuals diagnosed with BC. Currently, due to the absence of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor 2 (HER2) receptor, there is no hormone-based therapy for TNBC. In addition, there are still no FDA-approved targeted therapies for patients with TNBC. TNBC treatment is challenging owing to poor prognosis, tumor heterogeneity, chemotherapeutic side effects, the chance of metastasis, and multiple drug-resistance. Therefore, various bio-inspired tumor-homing nano systems responding to intra- and extra- cellular stimuli are an urgent need to treat TNBC patients who do not respond to current chemotherapy. In this review, intensive efforts have been made for exploring cell-membrane coated nanoparticles and immune cell-targeted nanoparticles (immunotherapy) to modulate the tumor microenvironment and deliver accurate amounts of therapeutic agents to TNBC without stimulating the immune system.
Collapse
Affiliation(s)
- Mahsa Keihan Shokooh
- Department of Pharmaceutics, College of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran;
| | | | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, Korea;
| |
Collapse
|