1
|
Karimova AF, Khalitova AR, Suezov R, Markov N, Mukhamedshina Y, Rizvanov AA, Huber M, Simon HU, Brichkina A. Immunometabolism of tumor-associated macrophages: A therapeutic perspective. Eur J Cancer 2025; 220:115332. [PMID: 40048925 DOI: 10.1016/j.ejca.2025.115332] [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: 12/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in the tumor microenvironment (TME), actively contributing to the formation of an immunosuppressive niche that fosters tumor progression. Consequently, there has been a growing interest in targeting TAMs as a promising avenue for cancer therapy. Recent advances in the field of immunometabolism have shed light on the influence of metabolic adaptations on macrophage physiology in the context of cancer. Here, we discuss the key metabolic pathways that shape the phenotypic diversity of macrophages. We place special emphasis on how metabolic reprogramming impacts the activation status of TAMs and their functions within the TME. Additionally, we explore alterations in TAM metabolism and their effects on phagocytosis, production of cytokines/chemokines and interaction with cytotoxic T and NK immune cells. Moreover, we examine the application of nanomedical approaches to target TAMs and assess the clinical significance of modulating the metabolism of TAMs as a strategy to develop new anti-cancer therapies. Taken together, in this comprehensive review article focusing on TAMs, we provide invaluable insights for the development of effective immunotherapeutic strategies and the enhancement of clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Adelya F Karimova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Adelya R Khalitova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Roman Suezov
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Yana Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Magdalena Huber
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany
| | - Hans-Uwe Simon
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
2
|
Li H, Liu K, Yang Z, Sun Q, Shang W, Li Y, Wang M, Yang Y, Liu H, Yin D, Shen W. Oncolytic polymer-mediated combretastatin A4 phosphate delivery for enhancing vascular disrupting therapy. Int J Pharm 2025; 674:125438. [PMID: 40058738 DOI: 10.1016/j.ijpharm.2025.125438] [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: 01/09/2025] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 04/19/2025]
Abstract
Although vascular disrupting agents (VDAs) can induce shutdown of blood flow and necrosis in the tumor core, eradicating tumor rim cells remains a significant challenge. Recently, researchers have developed various combination treatment strategies to improve the efficacy of VDAs. However, the aggravated hypoxic tumor microenvironment following vascular disruption limits the effectiveness of conventional therapeutic approaches. Here, we developed an ε-polylysine-derived oncolytic polymer (named OPAA) with membrane lytic activity. Its cytotoxic effect on tumor cells is largely unaffected by hypoxic conditions, as evidenced by the ratio of its IC50 value for 4 T1 cells under normoxic conditions to that under hypoxic conditions, which is 0.98. Subsequently, a pH-responsive combretastatin A4 phosphate disodium salt (CA4P)-loaded nanoparticle (OPAA@CA4P NPs) has been designed to efficiently deliver OPAA and CA4P to solid tumors. OPAA@CA4P NPs exhibited a prolonged serum half-life (t1/2 = 3.15 h) compared to CA4P (t1/2 = 0.31 h) and an enhanced tumor accumulation. In addition, CA4P can be responsively released within the tumor microenvironment, leading to necrosis in the tumor center. Concurrently, OPAA released from the nanoparticles eradicated the surviving cancer cells at the tumor periphery, thereby improving the overall therapeutic effect. Notably, compared to the CA4P + doxorubicin group (tumor suppression rates, TSR = 36.17 %), the OPAA@CA4P NPs group demonstrated superior therapeutic outcomes (TSR = 60.30 %). Overall, the introduction of oncolytic polymers provides new insights into the potential future applications of VDAs.
Collapse
Affiliation(s)
- Huihui Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Kang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wencui Shang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yunlong Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Mingjing Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China
| | - Hanmeng Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China; Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei 230021, China.
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), Hefei 230012, China.
| |
Collapse
|
3
|
Arandhara A, Bhuyan P, Das BK. Exploring lung cancer microenvironment: pathways and nanoparticle-based therapies. Discov Oncol 2025; 16:159. [PMID: 39934547 DOI: 10.1007/s12672-025-01902-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: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer stands out as a significant global health burden, with staggering incidence and mortality rates primarily linked to smoking and environmental carcinogens. The tumor microenvironment (TME) emerges as a critical determinant of cancer progression and treatment outcomes, comprising a complex interplay of cells, signaling molecules, and extracellular matrix. Through a comprehensive literature review, we elucidate current research trends and therapeutic prospects, aiming to advance our understanding of TME modulation strategies and their clinical implications for lung cancer treatment. Dysregulated immune responses within the TME can facilitate tumor evasion, limiting the efficacy of immune checkpoint inhibitors (ICI). Consequently, TME modulation strategies have become potential avenues to enhance therapeutic responses. However, conventional TME-targeted therapies often face challenges. In contrast, nanoparticle (NP)-based therapies offer promising prospects for improved drug delivery and reduced toxicity, leveraging the enhanced permeability and retention (EPR) effect. Despite NP design and delivery advancements, obstacles like poor tumor cell uptake and off-target effects persist, necessitating further optimization. This review underscores the pivotal role of TME in lung cancer management, emphasizing the synergistic potential of immunotherapy and nano-therapy.
Collapse
Affiliation(s)
- Arunabh Arandhara
- Assam Pharmacy Institute, Titabar, Amgurikhat, Jorhat, Assam, 785632, India
| | - Pallabi Bhuyan
- School of Pharmacy, The Assam Kaziranga University, Koraikhowa, Jorhat, Assam, 785006, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam, 781017, India.
| |
Collapse
|
4
|
Shao Y, Han S, Hou Z, Yang C, Zhao Y. Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon 2024; 10:e36839. [PMID: 39281573 PMCID: PMC11401039 DOI: 10.1016/j.heliyon.2024.e36839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Tumor-associated macrophages play an important role in the tumor immune microenvironment, and regulating the function of tumor-associated macrophages has important therapeutic potential in tumor therapy. Mature macrophages could migrate to the tumor microenvironment, influencing multiple factors such as tumor cell proliferation, invasion, metastasis, extracellular matrix remodeling, immune suppression, and drug resistance. As a major component of the tumor microenvironment, tumor-associated macrophages crosstalk with other immune cells. Currently, tumor-associated macrophages have garnered considerable attention in tumor therapy, broadening the spectrum of drug selection to some extent, thereby aiding in mitigating the prevailing clinical drug resistance dilemma. This article summarizes the recent advances in tumor-associated macrophages concerning immunology, drug targeting mechanisms for tumor-associated macrophages treatment, new developments, and existing challenges, offering insights for future therapeutic approaches. In addition, this paper summarized the impact of tumor-associated macrophages on current clinical therapies, discussed the advantages and disadvantages of targeted tumor-associated macrophages therapy compared with existing tumor therapies, and predicted and discussed the future role of targeted tumor-associated macrophages therapy and the issues that need to be focused on.
Collapse
Affiliation(s)
- Yanchi Shao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Song Han
- The First Hospital of Jilin University, Changchun, China
| | - Zhenxin Hou
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chen Yang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
He Y, Xu H, Liu Y, Kempa S, Vechiatto C, Schmidt R, Yilmaz EY, Heidemann L, Schnorr J, Metzkow S, Schellenberger E, Häckel A, Patzak A, Müller DN, Savic LJ. The Effects of Hypoxia on the Immune-Metabolic Interplay in Liver Cancer. Biomolecules 2024; 14:1024. [PMID: 39199411 PMCID: PMC11352590 DOI: 10.3390/biom14081024] [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: 06/14/2024] [Revised: 07/28/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
M2-like macrophages promote tumor growth and cancer immune evasion. This study used an in vitro model to investigate how hypoxia and tumor metabolism affect macrophage polarization. Liver cancer cells (HepG2 and VX2) and macrophages (THP1) were cultured under hypoxic (0.1% O2) and normoxic (21% O2) conditions with varying glucose levels (2 g/L or 4.5 g/L). Viability assays and extracellular pH (pHe) measurements were conducted over 96 hours. Macrophages were exposed to the tumor-conditioned medium (TCM) from the cancer cells, and polarization was assessed using arginase and nitrite assays. GC-MS-based metabolic profiling quantified TCM meta-bolites and correlated them with M2 polarization. The results showed that pHe in TCMs decreased more under hypoxia than normoxia (p < 0.0001), independent of glucose levels. The arginase assay showed hypoxia significantly induced the M2 polarization of macrophages (control group: p = 0.0120,0.1%VX2-TCM group: p = 0.0149, 0.1%HepG2-TCM group: p < 0.0001, 0.1%VX2-TCMHG group: p = 0.0001, and 0.1%HepG2-TCMHG group: p < 0.0001). TCMs also induced M2 polarization under normoxic conditions, but the strongest M2 polarization occurred when both tumor cells and macrophages were incubated under hypoxia with high glucose levels. Metabolomics revealed that several metabolites, particularly lactate, were correlated with hypoxia and M2 polarization. Under normoxia, elevated 2-amino-butanoic acid (2A-BA) strongly correlated with M2 polarization. These findings suggest that targeting tumor hypoxia could mitigate immune evasion in liver tumors. Lactate drives acidity in hypoxic tumors, while 2A-BA could be a therapeutic target for overcoming immunosuppression in normoxic conditions.
Collapse
Affiliation(s)
- Yubei He
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Han Xu
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
| | - Yu Liu
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany; (S.K.); (C.V.)
| | - Carolina Vechiatto
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany; (S.K.); (C.V.)
| | - Robin Schmidt
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Emine Yaren Yilmaz
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Luisa Heidemann
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Jörg Schnorr
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
| | - Susanne Metzkow
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
| | - Eyk Schellenberger
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
| | - Akvile Häckel
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
| | - Andreas Patzak
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Dominik N. Müller
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany; (S.K.); (C.V.)
| | - Lynn Jeanette Savic
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, 13353 Berlin, Germany; (Y.H.); (H.X.); (Y.L.); (R.S.); (E.Y.Y.); (L.H.); (J.S.); (S.M.); (E.S.); (A.H.)
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany;
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
6
|
Xu H, Russell SN, Steiner K, O'Neill E, Jones KI. Targeting PI3K-gamma in myeloid driven tumour immune suppression: a systematic review and meta-analysis of the preclinical literature. Cancer Immunol Immunother 2024; 73:204. [PMID: 39105848 PMCID: PMC11303654 DOI: 10.1007/s00262-024-03779-2] [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: 12/07/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024]
Abstract
The intricate interplay between immune and stromal cells within the tumour microenvironment (TME) significantly influences tumour progression. Myeloid cells, including tumour-associated macrophages (TAMs), neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), contribute to immune suppression in the TME (Nakamura and Smyth in Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo and Ruffell in Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 ). This poses a significant challenge for novel immunotherapeutics that rely on host immunity to exert their effect. This systematic review explores the preclinical evidence surrounding the inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) as a strategy to reverse myeloid-driven immune suppression in solid tumours. EMBASE, MEDLINE, and PubMed databases were searched on 6 October 2022 using keyword and subject heading terms to capture relevant studies. The studies, focusing on PI3Kγ inhibition in animal models, were subjected to predefined inclusion and exclusion criteria. Extracted data included tumour growth kinetics, survival endpoints, and immunological responses which were meta-analysed. PRISMA and MOOSE guidelines were followed. A total of 36 studies covering 73 animal models were included in the review and meta-analysis. Tumour models covered breast, colorectal, lung, skin, pancreas, brain, liver, prostate, head and neck, soft tissue, gastric, and oral cancer. The predominant PI3Kγ inhibitors were IPI-549 and TG100-115, demonstrating favourable specificity for the gamma isoform. Combination therapies, often involving chemotherapy, radiotherapy, immune checkpoint inhibitors, biological agents, or vaccines, were explored in 81% of studies. Analysis of tumour growth kinetics revealed a statistically significant though heterogeneous response to PI3Kγ monotherapy, whereas the tumour growth in combination treated groups were more consistently reduced. Survival analysis showed a pronounced increase in median overall survival with combination therapy. This systematic review provides a comprehensive analysis of preclinical studies investigating PI3Kγ inhibition in myeloid-driven tumour immune suppression. The identified studies underscore the potential of PI3Kγ inhibition in reshaping the TME by modulating myeloid cell functions. The combination of PI3Kγ inhibition with other therapeutic modalities demonstrated enhanced antitumour effects, suggesting a synergistic approach to overcome immune suppression. These findings support the potential of PI3Kγ-targeted therapies, particularly in combination regimens, as a promising avenue for future clinical exploration in diverse solid tumour types.
Collapse
Affiliation(s)
- Haonan Xu
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Keaton Ian Jones
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Salmaninejad A, Layeghi SM, Falakian Z, Golestani S, Kobravi S, Talebi S, Yousefi M. An update to experimental and clinical aspects of tumor-associated macrophages in cancer development: hopes and pitfalls. Clin Exp Med 2024; 24:156. [PMID: 39003350 PMCID: PMC11246281 DOI: 10.1007/s10238-024-01417-w] [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: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant tumor-infiltrating stromal cells, and their normal function in tumor microenvironment (TME) is to suppress tumor cells by producing cytokines which trigger both direct cell cytotoxicity and antibody-mediated immune response. However, upon prolonged exposure to TME, the classical function of these so-called M1-type TAMs can be converted to another type, "M2-type," which are recruited by tumor cells so that they promote tumor growth and metastasis. This is the reason why the accumulation of TAMs in TME is correlated with poor prognosis in cancer patients. Both M1- and M2-types have high degree of plasticity, and M2-type cells can be reprogrammed to M1-type for therapeutic purposes. This characteristic introduces TAMs as promising target for developing novel cancer treatments. In addition, inhibition of M2-type cells and blocking their recruitment in TME, as well as their depletion by inducing apoptosis, are other approaches for effective immunotherapy of cancer. In this review, we summarize the potential of TAMs to be targeted for cancer immunotherapy and provide an up-to-date about novel strategies for targeting TAMs.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sepideh Mehrpour Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Falakian
- Department of Laboratory Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Shahin Golestani
- Department of Ophthalmology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Tehran Azad University, Tehran, Iran
| | - Samaneh Talebi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
8
|
Li S, Sheng J, Zhang D, Qin H. Targeting tumor-associated macrophages to reverse antitumor drug resistance. Aging (Albany NY) 2024; 16:10165-10196. [PMID: 38787372 PMCID: PMC11210230 DOI: 10.18632/aging.205858] [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: 11/29/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Currently, antitumor drugs show limited clinical outcomes, mainly due to adaptive resistance. Clinical evidence has highlighted the importance of the tumor microenvironment (TME) and tumor-associated macrophages (TAMs) in tumor response to conventional antitumor drugs. Preclinical studies show that TAMs following antitumor agent can be reprogrammed to an immunosuppressive phenotype and proangiogenic activities through different mechanisms, mediating drug resistance and poor prognosis. Potential extrinsic inhibitors targeting TAMs repolarize to an M1-like phenotype or downregulate proangiogenic function, enhancing therapeutic efficacy of anti-tumor therapy. Moreover, pharmacological modulation of macrophages that restore the immune stimulatory characteristics is useful to reshaping the tumor microenvironment, thus further limiting tumor growth. This review aims to introduce macrophage response in tumor therapy and provide a potential therapeutic combination strategy of TAM-targeting immunomodulation with conventional antitumor drugs.
Collapse
Affiliation(s)
- Sheng Li
- The Second Hospital of Jilin University, Changchun, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Dan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Hanjiao Qin
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Chen J, Yin M, Yang C, Wang K, Ma L, Yu H, Huang Y, Liu F, Tang Z. Therapeutic effects and underlying mechanism of poly (L-glutamic acid)- g-methoxy poly (ethylene glycol)/combretastatin A4/BLZ945 nanoparticles on Renca renal carcinoma. Front Bioeng Biotechnol 2024; 12:1336692. [PMID: 38375454 PMCID: PMC10875097 DOI: 10.3389/fbioe.2024.1336692] [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: 11/11/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Introduction: The prognosis of advanced renal carcinoma is not ideal, necessitating the exploration of novel treatment strategies. Poly(L-glutamic acid)-g-methoxy poly(ethylene glycol)/Combretastatin A4 (CA4)/BLZ945 nanoparticles (CB-NPs) possess the dual capability of CA4 (targeting blood vessels to induce tumor necrosis) and BLZ945 (inducing M2 macrophage apoptosis), thereby inhibiting tumor growth. Methods: Here, the therapeutic effects and underlying mechanism was explored by CCK-8 cytotoxicity experiment, transwell cell invasion and migration experiment, H&E, western blot analysis, immunohistochemistry, flow cytometry, and other techniques. Results: These results demonstrated that CB-NPs could inhibit the growth of Renca cells and subcutaneous tumors in mice, with an impressive tumor inhibition rate of 88.0%. Results suggested that CB-NPs can induce necrosis in renal carcinoma cells and tissues, downregulate VEGFA expression, promote renal carcinoma cell apoptosis, and reduce the polarization of M2 macrophages. Discussion: These findings offer innovative perspectives for the treatment of advanced renal carcinoma.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Min Yin
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Chenguang Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Kun Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yue Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Feng Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
10
|
Lu C, Liu Y, Miao L, Kong X, Li H, Chen H, Zhao X, Zhang B, Cui X. Research progress on the role of tumor‑associated macrophages in tumor development and their use as molecular targets (Review). Int J Oncol 2024; 64:11. [PMID: 38063203 PMCID: PMC10734668 DOI: 10.3892/ijo.2023.5599] [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: 10/03/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The tumor microenvironment (TME) is a complex system composed mainly of tumor cells, mesenchymal cells and immune cells. Macrophages, also known as tumor‑associated macrophages (TAMs), among innate immune cells, are some of the most abundant components of the TME. They may influence tumor growth and metastasis through interactions with other cell populations in the TME and have been associated with poor prognosis in a variety of tumors. Therefore, a better understanding of the role of TAMs in the TME may provide new insight into tumor therapy. In the present review, the origin and classification of TAMs in the TME were outlined and their polarization and dual effects on tumor cells, as well as emerging strategies for cancer therapies targeting TAMs, were discussed.
Collapse
Affiliation(s)
- Chenglin Lu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Ying Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 860411, P.R. China
| | - Linxuan Miao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Xiangle Kong
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Huili Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Haoran Chen
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Xu Zhao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 860411, P.R. China
| |
Collapse
|
11
|
Zhao M, Cheng X, Shao P, Dong Y, Wu Y, Xiao L, Cui Z, Sun X, Gao C, Chen J, Huang Z, Zhang J. Bacterial protoplast-derived nanovesicles carrying CRISPR-Cas9 tools re-educate tumor-associated macrophages for enhanced cancer immunotherapy. Nat Commun 2024; 15:950. [PMID: 38296939 PMCID: PMC10830495 DOI: 10.1038/s41467-024-44941-9] [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: 03/23/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
The CRISPR-Cas9 system offers substantial potential for cancer therapy by enabling precise manipulation of key genes involved in tumorigenesis and immune response. Despite its promise, the system faces critical challenges, including the preservation of cell viability post-editing and ensuring safe in vivo delivery. To address these issues, this study develops an in vivo CRISPR-Cas9 system targeting tumor-associated macrophages (TAMs). We employ bacterial protoplast-derived nanovesicles (NVs) modified with pH-responsive PEG-conjugated phospholipid derivatives and galactosamine-conjugated phospholipid derivatives tailored for TAM targeting. Utilizing plasmid-transformed E. coli protoplasts as production platforms, we successfully load NVs with two key components: a Cas9-sgRNA ribonucleoprotein targeting Pik3cg, a pivotal molecular switch of macrophage polarization, and bacterial CpG-rich DNA fragments, acting as potent TLR9 ligands. This NV-based, self-assembly approach shows promise for scalable clinical production. Our strategy remodels the tumor microenvironment by stabilizing an M1-like phenotype in TAMs, thus inhibiting tumor growth in female mice. This in vivo CRISPR-Cas9 technology opens avenues for cancer immunotherapy, overcoming challenges related to cell viability and safe, precise in vivo delivery.
Collapse
Affiliation(s)
- Mingming Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xiaohui Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Pingwen Shao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Yao Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Yongjie Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Lin Xiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Zhiying Cui
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xuedi Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Chuancheng Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
- State Key Laboratory of Analytical Chemistry for Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
12
|
Qian Y, Yin Y, Zheng X, Liu Z, Wang X. Metabolic regulation of tumor-associated macrophage heterogeneity: insights into the tumor microenvironment and immunotherapeutic opportunities. Biomark Res 2024; 12:1. [PMID: 38185636 PMCID: PMC10773124 DOI: 10.1186/s40364-023-00549-7] [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: 10/01/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a heterogeneous population that play diverse functions in tumors. Their identity is determined not only by intrinsic factors, such as origins and transcription factors, but also by external signals from the tumor microenvironment (TME), such as inflammatory signals and metabolic reprogramming. Metabolic reprogramming has rendered TAM to exhibit a spectrum of activities ranging from pro-tumorigenic to anti-tumorigenic, closely associated with tumor progression and clinical prognosis. This review implicates the diversity of TAM phenotypes and functions, how this heterogeneity has been re-evaluated with the advent of single-cell technologies, and the impact of TME metabolic reprogramming on TAMs. We also review current therapies targeting TAM metabolism and offer new insights for TAM-dependent anti-tumor immunotherapy by focusing on the critical role of different metabolic programs in TAMs.
Collapse
Affiliation(s)
- Yujing Qian
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhaoyuan Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
13
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
14
|
Guo D, Huang Y, Wang K, Yang C, Ma L, Zhang Y, Yu H, Cui M, Tang Z. Preparation and Characterization Evaluation of Poly(L-Glutamic Acid)- g-Methoxy Poly(Ethylene Glycol)/Combretastatin A4/BLZ945 Nanoparticles for Cervical Cancer Therapy. Int J Nanomedicine 2023; 18:6901-6914. [PMID: 38026524 PMCID: PMC10676729 DOI: 10.2147/ijn.s441131] [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: 09/19/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Cervical cancer (CC) is a highly vascularized tumor with abundant abnormal blood vessel, which could be targeted by therapeutic strategies. Poly(L-glutamic acid)-g-methoxy poly(ethylene glycol)/combretastatin A4 (CA4)/BLZ945 nanoparticles (CB-NPs) have shown great potential as nano vascular disrupting agents (VDAs) in the realm of synergistic cancer therapy. Methods In this study, we investigated the nanocharacteristics of CB-NPs, focusing on active pharmaceutical ingredients (API), as well as lyophilized samples combining API with protective agents (PAs). The in vivo efficacy of final sample (API + PAs) was evaluated. Results The assembled sphere of API with complex core and thin-shell structure was confirmed. PAs were found to significantly influence in vivo efficacy. Collaborative efforts between API and PAs, namely mannitol and lactose, resulted in the most promising lyophilized sample, ie, the final sample (FS2) for CC therapy. Impressively, FS2 demonstrated an exceptional 100% cure rate on the CC U14-bearing mice model. Conclusion FS2 has provided significant insights for cervical cancer therapy. It is also crucial to develop a comprehensive evaluation strategy for the formulation of nanomedicine, which has the potential to serve as a guideline for future clinical trials.
Collapse
Affiliation(s)
- Dongmei Guo
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin Province, 130041, People’s Republic of China
| | - Yue Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Kun Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Chenguang Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Manhua Cui
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin Province, 130041, People’s Republic of China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| |
Collapse
|
15
|
Roshan MK, Afshari AR, Mirzavi F, Mousavi SH, Soukhtanloo M. Combretastatin A-4 suppresses the invasive and metastatic behavior of glioma cells and induces apoptosis in them: in-vitro study. Med Oncol 2023; 40:331. [PMID: 37838642 DOI: 10.1007/s12032-023-02197-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/16/2023]
Abstract
The most common primary brain malignancy, glioblastoma multiforme, is tremendously resistant to conventional treatments due to its potency for metastasis to surrounding brain tissue. Temozolomide is a chemotherapeutic agent that currently is administrated during the treatment procedure. Studies have attempted to investigate new agents with higher effectiveness and fewer side effects. Combretastatin A-4 (CA-4), a natural compound derived from Combretum caffrum, has been recently considered for its potent antitumor activities in a wide variety of preclinical solid tumor models. Our findings have shown that CA-4 exerts potent anti-proliferative and apoptotic effects on glioma cells, and ROS generation may be involved in these cellular events. CA-4 has imposed G2 arrest in U-87 cells. We also observed that CA-4 significantly reduced the migration and invasion capability of U-87 cells. Furthermore, the gene expression and enzyme activity of MMP-2 and MMP-9 were significantly inhibited in the presence of CA-4. We also observed a considerable decrease in PI3K and Akt protein expression following treatment with CA-4. In conclusion, our findings showed significant apoptogenic and anti-metastatic effects of CA-4 on glioma cells and also suggested that the PI3K/Akt/MMP-2/-9 and also ROS pathway might play roles in these cellular events.
Collapse
Affiliation(s)
- Mostafa Karimi Roshan
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Hadi Mousavi
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Liang Y, He J, Chen X, Yin L, Yuan Q, Zeng Q, Zu X, Shen Y. The emerging roles of metabolism in the crosstalk between breast cancer cells and tumor-associated macrophages. Int J Biol Sci 2023; 19:4915-4930. [PMID: 37781517 PMCID: PMC10539698 DOI: 10.7150/ijbs.86039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
Breast cancer is the most common cancer affecting women worldwide. Investigating metabolism in breast cancer may accelerate the exploitation of new therapeutic options for immunotherapies. Metabolic reprogramming can confer breast cancer cells (BCCs) with a survival advantage in the tumor microenvironment (TME) and metabolic alterations in breast cancer, and the corresponding metabolic byproducts can affect the function of tumor-associated macrophages (TAMs). Additionally, TAMs undergo metabolic reprogramming in response to signals present in the TME, which can affect their function and breast cancer progression. Here, we review the metabolic crosstalk between BCCs and TAMs in terms of glucose, lipids, amino acids, iron, and adenosine metabolism. Summaries of inhibitors that target metabolism-related processes in BCCs or TAMs within breast cancer have also served as valuable inspiration for novel therapeutic approaches in the fight against this disease. This review provides new perspectives on targeted anticancer therapies for breast cancer that combine immunity with metabolism.
Collapse
Affiliation(s)
- Yuxin Liang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguang Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiong Yuan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qiting Zeng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
17
|
Gu X, Zhang R, Sun Y, Ai X, Wang Y, Lyu Y, Wang X, Wu Y, Wang Z, Feng N, Liu Y. Oral membrane-biomimetic nanoparticles for enhanced endocytosis and regulation of tumor-associated macrophage. J Nanobiotechnology 2023; 21:206. [PMID: 37403048 DOI: 10.1186/s12951-023-01949-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Enterocyte uptake with high binding efficiency and minor endogenous interference remains a challenge in oral nanocarrier delivery. Enterocyte membrane-biomimetic lipids may universally cooperate with endogenous phosphatidyl choline via a biorthogonal group. In this study, we developed a sophorolipid-associated membrane-biomimetic choline phosphate-poly(lactic-co-glycolic) acid hybrid nanoparticle (SDPN). Aided by physical stability in the gastrointestinal tract and rapid mucus diffusion provided by association with sophorolipid, these nanoparticles show improved endocytosis, driven by dipalmitoyl choline phosphate-phosphatidyl choline interaction as well as its optimized membrane fluidity and rigidity. Luteolin- and silibinin-co-loaded with SDPN alleviated breast cancer metastasis in 4T1 tumor-bearing mice by regulating the conversion of tumor-associated M2 macrophages into the M1 phenotype and reducing the proportion of the M2-phenotype through co-action on STAT3 and HIF-1α. In addition, SDPN reduces angiogenesis and regulates the matrix barrier in the tumor microenvironment. In conclusion, this membrane-biomimetic strategy is promising for improving the enterocyte uptake of oral SDPN and shows potential to alleviate breast cancer metastasis.
Collapse
Affiliation(s)
- Xiaoyan Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Rongguang Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Yingwei Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Xinyi Ai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Yu Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Yaqi Lyu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yihan Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Zhi Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China
| | - Nianping Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China.
| | - Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, P R China.
| |
Collapse
|
18
|
Xiao L, Wang Q, Peng H. Tumor-associated macrophages: new insights on their metabolic regulation and their influence in cancer immunotherapy. Front Immunol 2023; 14:1157291. [PMID: 37426676 PMCID: PMC10325569 DOI: 10.3389/fimmu.2023.1157291] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are a dynamic and heterogeneous cell population of the tumor microenvironment (TME) that plays an essential role in tumor formation and progression. Cancer cells have a high metabolic demand for their rapid proliferation, survival, and progression. A comprehensive interpretation of pro-tumoral and antitumoral metabolic changes in TAMs is crucial for comprehending immune evasion mechanisms in cancer. The metabolic reprogramming of TAMs is a novel method for enhancing their antitumor effects. In this review, we provide an overview of the recent research on metabolic alterations of TAMs caused by TME, focusing primarily on glucose, amino acid, and fatty acid metabolism. In addition, this review discusses antitumor immunotherapies that influence the activity of TAMs by limiting their recruitment, triggering their depletion, and re-educate them, as well as metabolic profiles leading to an antitumoral phenotype. We highlighted the metabolic modulational roles of TAMs and their potential to enhance immunotherapy for cancer.
Collapse
Affiliation(s)
- Li Xiao
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Wang
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongling Peng
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Khan SU, Khan MU, Azhar Ud Din M, Khan IM, Khan MI, Bungau S, Hassan SSU. Reprogramming tumor-associated macrophages as a unique approach to target tumor immunotherapy. Front Immunol 2023; 14:1166487. [PMID: 37138860 PMCID: PMC10149956 DOI: 10.3389/fimmu.2023.1166487] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
In the last ten years, it has become increasingly clear that tumor-infiltrating myeloid cells drive not just carcinogenesis via cancer-related inflammatory processes, but also tumor development, invasion, and metastasis. Tumor-associated macrophages (TAMs) in particular are the most common kind of leucocyte in many malignancies and play a crucial role in establishing a favorable microenvironment for tumor cells. Tumor-associated macrophage (TAM) is vital as the primary immune cell subset in the tumor microenvironment (TME).In order to proliferate and spread to new locations, tumors need to be able to hide from the immune system by creating an immune-suppressive environment. Because of the existence of pro-tumoral TAMs, conventional therapies like chemotherapy and radiotherapy often fail to restrain cancer growth. These cells are also to blame for the failure of innovative immunotherapies premised on immune-checkpoint suppression. Understanding the series of metabolic changes and functional plasticity experienced by TAMs in the complex TME will help to use TAMs as a target for tumor immunotherapy and develop more effective tumor treatment strategies. This review summarizes the latest research on the TAMs functional status, metabolic changes and focuses on the targeted therapy in solid tumors.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Muhammad Azhar Ud Din
- Faculty of Pharmacy, Gomal University Dera Ismail Khan KPK, Dera Ismail Khan, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering Fuyang Normal University, Fuyang, China
| | - Muhammad Imran Khan
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Syed Shams ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Natural Product Chemistry, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Huang Y, Yang C, Lv J, Zhang Y, Wang K, Ma L, Liu Z, Yu H, Li M, Tang Z. Formula optimization and in vivo study of poly(L-glutamic acid)-g-methoxy poly(ethylene glycol)/combretastatin A4/BLZ945 nanoparticles for cancer therapy. Int J Pharm 2023; 636:122849. [PMID: 36933582 DOI: 10.1016/j.ijpharm.2023.122849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/18/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Poly(L-glutamic acid)-g-methoxy poly(ethylene glycol)/Combretastatin A4 (CA4)/BLZ945 nanoparticles (CB-NPs) have shown great potential in synergistic cancer therapy. However, it is still unclear how the nanoparticles' formula, such as injection dose, active agent ratio, and drug loading content, affects the side effects and in vivo efficacy of CB-NPs. In this study, a series of CB-NPs with different BLZ945/CA4 (B/C) ratios and drug loading contents were synthesized and evaluated on a hepatoma (H22) tumor-bearing mice model. The injection dose and B/C ratio were found to have a significant influence on the in vivo anticancer efficacy. The CB-NPs 20 with B/C weight ratio of 0.45/1, and total drug loading content (B + C) of 20.7 wt%, showed the highest potential for clinical application. Systematic pharmacokinetics, biodistribution, and in vivo efficacy evaluation for CB-NPs 20 have been finished, which may provide significant instruction for medicine screening and clinical application.
Collapse
Affiliation(s)
- Yue Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Chenguang Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jianlin Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Kun Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhilin Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
21
|
Xie J, Zhao X, Zhang P, Zhang Y, Cheng R, Zhong Z, Deng C. Codelivery of BCL2 and MCL1 Inhibitors Enabled by Phenylboronic Acid-Functionalized Polypeptide Nanovehicles for Synergetic and Potent Therapy of Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204866. [PMID: 36683178 PMCID: PMC10015845 DOI: 10.1002/advs.202204866] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Acute myeloid leukemia (AML) is the most refractory hematologic malignancy characterized by acute onset, rapid progression, and high recurrence rate. Here, codelivery of BCL2 (ABT199) and MCL1 (TW37) inhibitors using phenylboronic acid-functionalized polypeptide nanovehicles to achieve synergetic and potent treatment of AML is adopted. Leveraging the dynamic boronic ester bonds, BN coordination, and π-π stacking, the nanovehicles reveal remarkably efficient and robust drug coencapsulation. ABT199 can induce a series of pro-apoptotic reactions by promoting the dissociation of the pro-apoptotic protein Bim from BCL2, while the released Bim is often captured by MCL1 protein overexpressed in AML. TW37 has a strong inhibitory ability to MCL1, thereby can restrain the depletion of Bim protein. Dual inhibitor-loaded nanoparticles (NPAT) reveal excellent stability, acid/enzyme/H2 O2 -triggered drug release, and significant cytotoxicity toward MOLM-13-Luc and MV-411 AML cells with low half maximal inhibitory concentrations of 1.15 and 7.45 ng mL-1 , respectively. In mice bearing MOLM-13-Luc or MV-411 AML cancer, NPAT reveal significant inhibition of tumor cell infiltration in bone marrow and main organs, potent suppression of tumor growth, and remarkably elevated mouse survival. With facile construction, varying drug combination, superior safety, synergetic efficacy, the phenylboronic acid-functionalized smart nanodrugs hold remarkable potential for AML treatment.
Collapse
Affiliation(s)
- Jiguo Xie
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Xiaofei Zhao
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Peng Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Yueyue Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Ru Cheng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Chao Deng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
22
|
Chen C, Zhang W, Lu SY, Wang J, Tan Y, Zhao S, Ouyang Y, Xu L, Zhou B, Yin X, Ran H, Liu H. Repolarizing tumor-associated macrophages by layered double hydroxide-based deacidification agent for tumor chemodynamic therapy and immunotherapy. Colloids Surf B Biointerfaces 2023; 223:113157. [PMID: 36716658 DOI: 10.1016/j.colsurfb.2023.113157] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/23/2023]
Abstract
Tumor-associated macrophages (TAMs)-mediated immunotherapy has attracted extensive attention in tumor elimination. However, the acidic tumor microenvironment (TME) severely limits the phenotype of TAMs to pro-tumoral M2 state, suppressing immune response efficacy against tumors. Herein, novel poly(acrylic acid) (PAA)-coated, doxorubicin (DOX)-loaded layered double hydroxide (LDH) nanosheets (NSs) were developed as deacidification agent to repolarize TAMs from pro-tumoral M2 to anti-tumoral M1 phenotype for tumor elimination through combined chemodynamic therapy and immunotherapy. When located in tumor regions, LDH-PAA@DOX NSs display good deacidification capacity to neutralize acidic TME, achieving the repolarization of TAMs to M1 phenotype and further activating CD8+ T cells. During the deacidification process, these NSs are acid-responsive and degrade to release Fe3+ and DOX. The former can be reduced to Fe2+ by intracellular glutathione, meanwhile disrupting the antioxidant defense system of tumor cells. The latter can damage tumor cells directly and further stimulate the production of hydrogen peroxide, providing abundant substrate for the Fenton reaction. Toxic hydroxyl radical is excessively produced through Fe2+-mediated Fenton reaction to cause intratumoral oxidative stress. In vivo data revealed that significant tumor elimination can be achieved under LDH-PAA@DOX treatment. This work not only provides a promising paradigm for neutralizing acidic TME using deacidification agent but also highlights the effectiveness of combined chemodynamic therapy and immunotherapy in tumor treatment.
Collapse
Affiliation(s)
- Chunmei Chen
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Shi-Yu Lu
- College of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Jingjing Wang
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yixin Tan
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Sheng Zhao
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yi Ouyang
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Luen Xu
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Benqing Zhou
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, China.
| | - Xuntao Yin
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.
| | - Hui Liu
- School of Materials and Energy, Southwest University, Chongqing 400715, China.
| |
Collapse
|
23
|
Zhan C, Jin Y, Xu X, Shao J, Jin C. Antitumor therapy for breast cancer: Focus on tumor-associated macrophages and nanosized drug delivery systems. Cancer Med 2023. [PMID: 36794651 DOI: 10.1002/cam4.5489] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/15/2022] [Accepted: 11/17/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND In breast cancer (BC), tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment and are closely related to poor prognosis. A growing number of studies have focused on the role of TAMs in BC progression and therapeutic strategies targeting TAMs. As an emerging treatment, the application of nanosized drug delivery systems (NDDSs) in the treatment of BC by targeting TAMs has attracted much attention. AIMS This review is to summarize the characteristics and treatment strategies targeting TAMs in BC and to clarify the applications of NDDSs targeting TAMs in the treatment of BC by targeting TAMs. MATERIALS & METHODS The existing results related to characteristics of TAMs in BC, BC treatment strategies by targeting TAMs, and the applications of NDDSs in these strategies are described. Through analyzing these results, the advantages and disadvantages of the treatment strategies using NDDSs are discussed, which could provide advices on designing NDDSs for BC treatment. RESULTS TAMs are one of the most prominent noncancer cell types in BC. TAMs not only promote angiogenesis, tumor growth and metastasis but also lead to therapeutic resistance and immunosuppression. Mainly four strategies have been used to target TAMs for BC therapy, which include depleting macrophages, blocking recruitment, reprogramming to attain an anti-tumor phenotype, and increasing phagocytosis. Since NDDSs can efficiently deliver drugs to TAMs with low toxicity, they are promising approaches for targeting TAMs in tumor therapy. NDDSs with various structures can deliver immunotherapeutic agents and nucleic acid therapeutics to TAMs. In addition, NDDSs can realize combination therapies. DISCUSSION TAMs play a critical role in the progression of BC. An increasing number of strategies have been proposed to regulate TAMs. Compared with free drugs, NDDSs targeting TAMs improve drug concentration, reduce toxicity and realize combination therapies. However, in order to achieve better therapeutic efficacy, there are still some disadvantages that need to be considered in the design of NDDSs. CONCLUSION TAMs play an important role in the progression of BC, and targeting TAMs is a promising strategy for BC therapy. In particular, NDDSs targeting TAMs have unique advantages and are potential treatments for BC.
Collapse
Affiliation(s)
- Cuiping Zhan
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinzhi Xu
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China.,Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiangbo Shao
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chunxiang Jin
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
Wang P, Li M, Zhou F, Yang Y, Yin X, Zhang XB, Song G. COF-based nanoreactors for click-activated prodrug delivery and precise anti-vascular therapy. Chem Commun (Camb) 2022; 58:11107-11110. [PMID: 36102676 DOI: 10.1039/d2cc03931a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein, we report a new click-activated prodrug, CA4V, and a bioorthogonal nanoreactor, CA4V/ZIF-90@TzCOF@Apt, which consists of a ZIF-90 core, tetrazine-based covalent organic framework (COF) shells and an aptamer polymer coating. When targeting a tumor, the acid-causing collapse of ZIF-90 initiates a nanoconfined bioorthogonal reaction in defined COF cages, which boosts the click efficiency of CA4V activation and therapeutic effects in vivo.
Collapse
Affiliation(s)
- Peng Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China. .,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Mili Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Fang Zhou
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China. .,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yue Yang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Xia Yin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Guosheng Song
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
25
|
Zhang Y, Kim I, Lu Y, Xu Y, Yu DG, Song W. Intelligent poly(l-histidine)-based nanovehicles for controlled drug delivery. J Control Release 2022; 349:963-982. [PMID: 35944751 DOI: 10.1016/j.jconrel.2022.08.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022]
Abstract
Stimuli-responsive drug delivery systems based on polymeric nanovehicles are among the most promising treatment regimens for malignant cancers. Such intelligent systems that release payloads in response to the physiological characteristics of tumor sites have several advantages over conventional drug carriers, offering, in particular, enhanced therapeutic effects and decreased toxicity. The tumor microenvironment (TME) is acidic, suggesting the potential of pH-responsive nanovehicles for enhancing treatment specificity and efficacy. The synthetic polypeptide poly(l-histidine) (PLH) is an appropriate candidate for the preparation of pH-responsive nanovehicles because the pKa of PLH (approximately 6.0) is close to the pH of the acidic TME. In addition, the pendent imidazole rings of PLH yield pH-dependent hydrophobic-to-hydrophilic phase transitions in the acidic TME, triggering the destabilization of nanovehicles and the subsequent release of encapsulated chemotherapeutic agents. Herein, we highlight the state-of-the-art design and construction of pH-responsive nanovehicles based on PLH and discuss the future challenges and perspectives of this fascinating biomaterial for targeted cancer treatment and "benchtop-to-clinic" translation.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China.
| | - Il Kim
- School of Chemical Engineering, Pusan National University, Busan 46241, Republic of Korea.
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, PR China.
| | - Wenliang Song
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, PR China.
| |
Collapse
|
26
|
Tang Y, Varyambath A, Ding Y, Chen B, Huang X, Zhang Y, Yu DG, Kim I, Song W. Porous organic polymers for drug delivery: hierarchical pore structures, variable morphologies, and biological properties. Biomater Sci 2022; 10:5369-5390. [PMID: 35861101 DOI: 10.1039/d2bm00719c] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Porous organic polymers have received considerable attention in recent years because of their applicability as biomaterials. In particular, their hierarchical pore structures, variable morphologies, and tunable biological properties make them suitable as drug-delivery systems. In this review, the synthetic and post forming/control methods including templated methods, template-free methods, mechanical methods, electrospun methods, and 3D printing methods for controlling the hierarchical structures and morphologies of porous organic polymers are discussed, and the different methods affecting their specific surface areas, hierarchical structures, and unique morphologies are highlighted in detail. In addition, we discuss their applications in drug encapsulation and the development of stimuli (pH, heat, light, and dual-stimuli)-responsive materials, focusing on their use for targeted drug release and as therapeutic agents. Finally, we present an outlook concerning the research directions and applications of porous polymer-based drug delivery systems.
Collapse
Affiliation(s)
- Yunxin Tang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China.
| | - Anuraj Varyambath
- BK21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Busan 609-735, Republic of Korea.
| | - Yuanchen Ding
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China.
| | - Bailiang Chen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China.
| | - Xinyi Huang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China.
| | - Yu Zhang
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, P. R. China.
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China.
| | - Il Kim
- BK21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Busan 609-735, Republic of Korea.
| | - Wenliang Song
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China. .,State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
27
|
Cao X, Lai SWT, Chen S, Wang S, Feng M. Targeting tumor-associated macrophages for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:61-108. [PMID: 35636930 DOI: 10.1016/bs.ircmb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant immune components in the tumor microenvironment and play a plethora of roles in regulating tumorigenesis. Therefore, the therapeutic targeting of TAMs has emerged as a new paradigm for immunotherapy of cancer. Herein, the review summarizes the origin, polarization, and function of TAMs in the progression of malignant diseases. The understanding of such knowledge leads to several distinct therapeutic strategies to manipulate TAMs to battle cancer, which include those to reduce TAM abundance, such as depleting TAMs or inhibiting their recruitment and differentiation, and those to harness or boost the anti-tumor activities of TAMs such as blocking phagocytosis checkpoints, inducing antibody-dependent cellular phagocytosis, and reprogramming TAM polarization. In addition, modulation of TAMs may reshape the tumor microenvironment and therefore synergize with other cancer therapeutics. Therefore, the rational combination of TAM-targeting therapeutics with conventional therapies including radiotherapy, chemotherapy, and other immunotherapies is also reviewed. Overall, targeting TAMs presents itself as a promising strategy to add to the growing repertoire of treatment approaches in the fight against cancer, and it is hopeful that these approaches currently being pioneered will serve to vastly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| | - Seigmund W T Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sadira Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
28
|
Yin R, Zhai X, Han H, Tong X, Li Y, Deng K. Characterizing the landscape of cervical squamous cell carcinoma immune microenvironment by integrating the single-cell transcriptomics and RNA-Seq. Immun Inflamm Dis 2022; 10:e608. [PMID: 35634956 PMCID: PMC9091987 DOI: 10.1002/iid3.608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/21/2022] Open
Abstract
Background Cervical squamous cell carcinoma (CSCC), caused by the infection of high‐risk human papillomavirus, is one of the most common malignancies in women worldwide. Methods RNA expression data, including those from the Cancer Genome Atlas, Gene Expression Omnibus, and Genotype‐Tissue Expression databases, were used to identify the expression of RNAs in normal and tumor tissue. Correlation analysis was performed to identify the immune‐related long noncoding RNAs (IRLs) and hypoxia‐related genes (IRHs) that can influence the activity of the immune system. Prognosis models of immune‐related RNAs (IRRs) were used to construct a coexpression network of the immune system. We identified the role of IRRs in immunotherapy by correlation analysis with immune checkpoint genes (ICGs). We then validated the expression data by integrating two single‐cell sequencing data sets of CSCC to identify the key immune features. Results In total, six immune‐related gene (IRG), four IRL, and five IRH signatures that can significantly influence the characteristics of the tumor immune microenvironment (TIME) were selected using machine learning methods. The expression level of ICGs was significantly upregulated in GZMB+CD8+ T‐cells and tumor‐associated macrophages (TAMs) in tumor tissues. TGFBI+ TAMs are a kind of blood‐derived monocyte‐derived M0‐like TAM linked to hypoxia and a poor prognosis. IFI30+ M1‐like TAMs participate in the process of immune‐regulation and showed a role in the promotion of CD8+ T‐cells and Type 1 T helper (Th1)/Th2 cells in the coexpression network, together with several IRLs, IRGs, and ICGs. Conclusions CD16+ monocyte‐derived IFI30+ TAMs participated in our coexpression network to regulate the TIME, showing the potential to be a novel immunotherapy target. The enrichment of M0‐like TAMs was associated with a worse prognosis in the high‐risk score group with IRH signatures. Remarkably, M0‐like TAMs in tumor tissues overexpressed TGFBI and were associated with several well‐known tumor‐proliferation pathways.
Collapse
Affiliation(s)
- Ruiling Yin
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiuming Zhai
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyan Han
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuedong Tong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Li
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kun Deng
- Department of Laboratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
29
|
Zhang Y, Song W, Lu Y, Xu Y, Wang C, Yu DG, Kim I. Recent Advances in Poly(α- L-glutamic acid)-Based Nanomaterials for Drug Delivery. Biomolecules 2022; 12:636. [PMID: 35625562 PMCID: PMC9138577 DOI: 10.3390/biom12050636] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
Poly(α-L-glutamic acid) (PGA) is a class of synthetic polypeptides composed of the monomeric unit α-L-glutamic acid. Owing to their biocompatibility, biodegradability, and non-immunogenicity, PGA-based nanomaterials have been elaborately designed for drug delivery systems. Relevant studies including the latest research results on PGA-based nanomaterials for drug delivery have been discussed in this work. The following related topics are summarized as: (1) a brief description of the synthetic strategies of PGAs; (2) an elaborated presentation of the evolving applications of PGA in the areas of drug delivery, including the rational design, precise fabrication, and biological evaluation; (3) a profound discussion on the further development of PGA-based nanomaterials in drug delivery. In summary, the unique structures and superior properties enables PGA-based nanomaterials to represent as an enormous potential in biomaterials-related drug delivery areas.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Wenliang Song
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea;
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Changping Wang
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Deng-Guang Yu
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Il Kim
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
30
|
Liu Z, Zhang Y, Shen N, Sun J, Tang Z, Chen X. Destruction of tumor vasculature by vascular disrupting agents in overcoming the limitation of EPR effect. Adv Drug Deliv Rev 2022; 183:114138. [PMID: 35143895 DOI: 10.1016/j.addr.2022.114138] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/27/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023]
Abstract
Nanomedicine greatly improves the efficiency in the delivery of antitumor drugs into the tumor, but insufficient tumoral penetration impairs the therapeutic efficacy of most nanomedicines. Vascular disrupting agent (VDA) nanomedicines are distributed around the tumor vessels due to the low tissue penetration in solid tumors, and the released drugs can selectively destroy immature tumor vessels and block the supply of oxygen and nutrients, leading to the internal necrosis of the tumors. VDAs can also improve the vascular permeability of the tumor, further increasing the extravasation of VDA nanomedicines in the tumor site, markedly reducing the dependence of nanomedicines on the enhanced permeability and retention effect (EPR effect). This review highlights the progress of VDA nanomedicines in recent years and their application in cancer therapy. First, the mechanisms of different VDAs are introduced. Subsequently, different strategies of delivering VDAs are described. Finally, multiple combination strategies with VDA nanomedicines in cancer therapy are described in detail.
Collapse
|
31
|
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: 47] [Impact Index Per Article: 15.7] [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
|
32
|
Analysis on the Clinical Effect of High-Dose Glucocorticoids Combined with Immunosuppressants on Patients with Myasthenia Gravis Undergoing Video-Assisted Thoracoscopic Surgery. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5854056. [PMID: 34912892 PMCID: PMC8668280 DOI: 10.1155/2021/5854056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022]
Abstract
Objective The purpose of the study was to investigate the clinical effect of high-dose glucocorticoids (GCS) combined with immunosuppressants on the treatment of myasthenia gravis (MG) with video-assisted thoracoscopic surgery (VATS). Methods A total of 106 MG patients admitted to the neurology department of our hospital from February 2016 to February 2020 were selected as the study subjects and divided into experimental group (n = 53) and control group (n = 53). The patients in the control group underwent VATS, while the patients in the experimental group were treated with high-dose GCS combined with immunosuppressants on the basis of VATS treatment. The clinical efficacy of different MG treatment methods was analyzed. Results No significant differences were observed in visual analogue score (VAS) at T1 between the two groups (P > 0.05), while VAS scores at T2, T3, and T4 in the experimental group were significantly lower than those in the control group (P < 0.001). In the experimental group, the overall response rate was significantly higher than the control group (P < 0.05). Cytotoxic T-lymphocyte-associated protein 4 (CTLA4) level in regulatory T (Treg) cells in experimental groups after treatment was significantly higher, compared to that in before treatment and the control group (P < 0.05). Similar results of each quantitative MG score were displayed in both groups after treatment, compared to before treatment and the control group (P < 0.05). Clinical performance of patients with lower incidence of adverse reactions in the experimental groups after treatment was significantly higher than those in the control group (P < 0.001). Conclusion GCS combined with immunosuppressants can effectively relieve patients' clinical symptoms and improve their quality of life, with significant clinical efficacy and high safety, which is worthy of application and promotion.
Collapse
|
33
|
Lopez-Yrigoyen M, Cassetta L, Pollard JW. Macrophage targeting in cancer. Ann N Y Acad Sci 2021; 1499:18-41. [PMID: 32445205 DOI: 10.1111/nyas.14377] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022]
Abstract
Tumorigenesis is not only determined by the intrinsic properties of cancer cells but also by their interactions with components of the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are among the most abundant immune cells in the TME. During initial stages of tumor development, macrophages can either directly promote antitumor responses by killing tumor cells or indirectly recruit and activate other immune cells. As genetic changes occur within the tumor or T helper 2 (TH 2) cells begin to dominate the TME, TAMs begin to exhibit an immunosuppressive protumor phenotype that promotes tumor progression, metastasis, and resistance to therapy. Thus, targeting TAMs has emerged as a strategy for cancer therapy. To date, TAM targeting strategies have focused on macrophage depletion and inhibition of their recruitment into the TME. However, these strategies have shown limited therapeutic efficacy, although trials are still underway with combination therapies. The fact that macrophages have the potential for antitumor activity has moved the TAM targeting field toward the development of TAM-reprogramming strategies to support this antitumor immune response. Here, we discuss the various roles of TAMs in cancer therapy and their immunosuppressive properties, as well as implications for emerging checkpoint inhibitor-based immunotherapies. We review state-of-the-art TAM-targeting strategies, focusing on current ones at the preclinical and clinical trial stages that aim to reprogram TAMs as an oncological therapy.
Collapse
Affiliation(s)
- Martha Lopez-Yrigoyen
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Luca Cassetta
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Berta GN, Di Scipio F, Yang Z, Oberto A, Abbadessa G, Romano F, Carere ME, Ceccarelli A, Hirsch E, Mognetti B. Chemical Oral Cancerogenesis Is Impaired in PI3Kγ Knockout and Kinase-Dead Mice. Cancers (Basel) 2021; 13:cancers13164211. [PMID: 34439365 PMCID: PMC8391366 DOI: 10.3390/cancers13164211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/08/2022] Open
Abstract
We investigated the role of PI3Kγ in oral carcinogenesis by using a murine model of oral squamous carcinoma generated by exposure to 4-nitroquinoline 1-oxide (4NQO) and the continuous human cancer cell line HSC-2 and Cal-27. PI3Kγ knockout (not expressing PI3Kγ), PI3Kγ kinase-dead (carrying a mutation in the PI3Kγ gene causing loss of kinase activity) and wild-type (WT) C57Bl/6 mice were administered 4NQO via drinking water to induce oral carcinomas. At sacrifice, lesions were histologically examined and stained for prognostic tumoral markers (EGFR, Neu, cKit, Ki67) and inflammatory infiltrate (CD3, CD4, CD8, CD19 and CD68). Prevalence and incidence of preneoplastic and exophytic lesions were significantly and similarly delayed in both transgenic mice versus the control. The expression of prognostic markers, as well as CD19+ and CD68+ cells, was higher in WT, while T lymphocytes were more abundant in tongues isolated from transgenic mice. HSC-2 and Cal-27 cells were cultured in the presence of the specific PI3Kγ-inhibitor (IPI-549) which significantly impaired cell vitality in a dose-dependent manner, as shown by the MTT test. Here, we highlighted two different mechanisms, namely the modulation of the tumor-infiltrating cells and the direct inhibition of cancer-cell proliferation, which might impair oral cancerogenesis in the absence/inhibition of PI3Kγ.
Collapse
Affiliation(s)
- Giovanni Nicolao Berta
- Department of Clinical and Biological Science, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy; (F.D.S.); (G.A.); (M.E.C.); (A.C.)
- Correspondence: (G.N.B.); (B.M.); Tel.: +39-011-670-5446 (G.N.B.); +39-011-670-4518 (B.M.)
| | - Federica Di Scipio
- Department of Clinical and Biological Science, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy; (F.D.S.); (G.A.); (M.E.C.); (A.C.)
| | - Zhiqian Yang
- Scientific Research Center, First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Guangzhou 510080, China;
| | - Alessandra Oberto
- Department of Neuroscience, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy;
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, Regione Gonzole 10, 10043 Orbassano, TO, Italy
| | - Giuliana Abbadessa
- Department of Clinical and Biological Science, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy; (F.D.S.); (G.A.); (M.E.C.); (A.C.)
| | - Federica Romano
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, 10126 Turin, Italy;
| | - Maria Elisabetta Carere
- Department of Clinical and Biological Science, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy; (F.D.S.); (G.A.); (M.E.C.); (A.C.)
| | - Adriano Ceccarelli
- Department of Clinical and Biological Science, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy; (F.D.S.); (G.A.); (M.E.C.); (A.C.)
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, Regione Gonzole 10, 10043 Orbassano, TO, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| | - Barbara Mognetti
- Department of Life Science and System Biology, University of Turin, Via Accademia Albertina 13, 10123 Turin, Italy
- Correspondence: (G.N.B.); (B.M.); Tel.: +39-011-670-5446 (G.N.B.); +39-011-670-4518 (B.M.)
| |
Collapse
|
35
|
Prognostic significance of the systemic immune-inflammation index in pancreatic carcinoma patients: A meta-analysis. Biosci Rep 2021; 41:229290. [PMID: 34286342 PMCID: PMC8329648 DOI: 10.1042/bsr20204401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Systemic immune-inflammation index (SII) is a prognostic indicator for several malignancies, including pancreatic carcinoma; however, there is no consensus on its significance. In the current study, a systematic meta-analysis was used to explore the correlation between SII and prognosis in pancreatic carcinoma patients. Methods: PubMed, Embase and Cochrane Library databases were screened from inception to May 2020. Studies describing the prognostic role of SII in pancreatic carcinoma were then retrieved. The pooled hazard ratio (HR) and 95% confidence interval (CI) was calculated using random- or fixed-effects models to determine the correlation between SII and prognosis. Results: A total of four studies, comprising 1749 patients, met the inclusion criteria of the study and were therefore included in this meta-analysis. The meta-analysis showed that high SII indicated was correlated with worse overall survival (OS) in patients with pancreatic carcinoma (HR: 1.43, 95% CI: 1.24–1.65, P<0.001). These findings were validated through subgroup analyses, stratified by the American Joint Committee on Cancer (AJCC) stage. In addition, patients with high SII showed poorer cancer-specific survival (HR: 2.32, 95% CI: 1.55–3.48, P<0.001). However, analysis showed no significant correlations between SII and disease-free and relapse-free survival (RFS). Conclusion: These findings indicate that SII is a potential non-invasive and a promising tool for predicting clinical outcomes of pancreatic carcinoma patients. However, the current research did not explore whether neoadjuvant therapy has an effect on the prognostic value of SII. Further studies using adequate designs and larger sample sizes are required to validate these findings.
Collapse
|
36
|
Feng Y, Xie X, Zhang H, Su Q, Yang G, Wei X, Li N, Li T, Qin X, Li S, Wu C, Zheng C, Zhu J, You F, Wang G, Yang H, Liu Y. Multistage-responsive nanovehicle to improve tumor penetration for dual-modality imaging-guided photodynamic-immunotherapy. Biomaterials 2021; 275:120990. [PMID: 34186239 DOI: 10.1016/j.biomaterials.2021.120990] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/12/2021] [Accepted: 06/21/2021] [Indexed: 01/10/2023]
Abstract
The exploration of an intelligent multifunctional imaging-guided therapeutic platform is of great significance because of its ideal delivery efficiency and controlled release. In this work, a tumor microenvironment (TME)-responsive nanocarrier (denoted as MB@MSP) is designed for on-demand, sequentially release of a short D-peptide antagonist of programmed cell death-ligand 1 (named as PDPPA-1) and a photosensitizer methylene blue (MB). Fe3O4-Au located in the core of MB@MSP is used as a magnetic resonance imaging and micro-computed tomography imaging contrast agent for noninvasive diagnosis of solid tumors and simultaneous monitoring of drug delivery. The PDPPA-1 coated on MB@MSP can be shed due to the cleavage of the peptide substrate by matrix metalloproteinase-2 (MMP-2) that is highly expressed in the tumor stroma, and disulfide bonding is further broken when it encounters high levels of glutathione (GSH) in TME, which finally leads to significant size reduction and charge-reversal. These transitions facilitate penetration and uptake of nanocarriers against tumors. Noticeably, the released PDPPA-1 can block the immune checkpoint to create an environment that favors the activation of cytotoxic T lymphocytes and augment the antitumor immune response elicited by photodynamic therapy, thus significantly improving therapeutic outcomes. Studies of the underlying mechanisms suggest that the designed MMP-2 and GSH-sensitive delivery system not only induce apoptosis of tumor cells but also modulate the immunosuppressive tumor microenvironment to eventually augment the suppression tumor metastasis effect of CD8+ cytotoxic T cells. Overall, the visualization of the therapeutic processes with comprehensive information renders MB@MSP an intriguing platform to realize the combined treatment of metastatic tumors.
Collapse
Affiliation(s)
- Yi Feng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Xiaoxue Xie
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Hanxi Zhang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Qingqing Su
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Geng Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Xiaodan Wei
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Ningxi Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, 400044, PR China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Jie Zhu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, 400044, PR China.
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China.
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China.
| |
Collapse
|
37
|
Zhang Y, He P, Zhang P, Yi X, Xiao C, Chen X. Polypeptides-Drug Conjugates for Anticancer Therapy. Adv Healthc Mater 2021; 10:e2001974. [PMID: 33929786 DOI: 10.1002/adhm.202001974] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/30/2021] [Indexed: 12/15/2022]
Abstract
Polypeptides are an important class of biodegradable polymers that have been widely used in drug delivery field. Owing to the controllable synthesis and robust side chain-functionalization ability, polypeptides have long been ideal candidates for conjugation with anticancer drugs. The chemical conjugation of anticancer drugs with polypeptides, termed polypeptides-drug conjugates, has demonstrated several advantages in improving pharmacokinetics, enhancing drug targeting, and controlling drug release, thereby leading to enhanced therapeutic outcomes with reduced side toxicities. This review focuses on the recent advances in the design and preparation of polypeptides-drug conjugates for enhanced anticancer therapy. Strategies for conjugation of different types of drugs, including small-molecule chemotherapeutic drugs, proteins, vascular disrupting agents, and gas molecules, onto polypeptides backbone are summarized. Finally, the challenges and future perspectives on the development of innovative polypeptides-drug conjugates for clinical cancer treatment are also presented.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Pan He
- School of Materials Science and Engineering Changchun University of Science and Technology Changchun 130022 P. R. China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| |
Collapse
|
38
|
Zhang Y, Liu X, Wang X, He P, Xiao C, Yu H, Chen X. Nanoparticles Composed of PEGylated Alternating Copolymer-Combretastatin A4 Conjugate for Cancer Therapy. Macromol Biosci 2021; 21:e2100077. [PMID: 34031970 DOI: 10.1002/mabi.202100077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Indexed: 12/18/2022]
Abstract
Chemotherapy using vascular targeting agents is an emerging new approach for cancer therapy. Combretastatin A4 (CA4) is a leading vascular-disrupting agent that targets the tumor blood vasculature for clinical tumor elimination. However, the extremely poor water solubility of CA4 hinders its biomedical applications. In this study, nanoparticles composed of novel PEGylated alternating copolymer-CA4 conjugates are designed to improve the therapeutic efficiency of CA4. First, an alternating copolymer with an alkene-pendant is synthesized by mPEG-OH-initiated ring-opening copolymerization. Then, side carboxyl groups are introduced by a thio-ene "click" chemical reaction, followed with CA4 conjugation through the Yamaguchi-reaction, resulting in the target copolymer, mPEG-b-P(PA-alt-GCA4). Interestingly, the polymer-drug conjugates can self-assemble into nanoparticles with an average diameter of 55.6 nm. The in vitro drug release and cytotoxicity of the obtained CA4-NPs toward 4T1 cells are investigated. Finally, the antitumor efficiency is evaluated in a 4T1-tumor bearing murine model. The in vivo test results demonstrate that CA4-NPs inhibited tumor growth much more efficiently at doses of 30 and 60 mg kg-1 , compared with the control group.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xinming Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xueping Wang
- School of Materials Science and Engineering, Changchun University of Science and Technology, Changchun, 130022, P. R. China
| | - Pan He
- School of Materials Science and Engineering, Changchun University of Science and Technology, Changchun, 130022, P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
39
|
Huang X, Zhang F, He D, Ji X, Gao J, Liu W, Wang Y, Liu Q, Xin T. Immune-Related Gene SERPINE1 Is a Novel Biomarker for Diffuse Lower-Grade Gliomas via Large-Scale Analysis. Front Oncol 2021; 11:646060. [PMID: 34094933 PMCID: PMC8173178 DOI: 10.3389/fonc.2021.646060] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Background Glioma is one of the highly fatal primary tumors in the central nervous system. As a major component of tumor microenvironment (TME), immune cell has been proved to play a critical role in the progression and prognosis of the diffuse lower-grade gliomas (LGGs). This study aims to screen the key immune-related factors of LGGs by investigating the TCGA database. Methods The RNA-sequencing data of 508 LGG patients were downloaded in the TCGA database. ESTIMATE algorithm was utilized to calculate the stromal, immune, and ESTIMATE scores, based on which, the differentially expressed genes (DEGs) were analyzed by using “limma” package. Cox regression analysis and the cytoHubba plugin of Cytoscape software were subsequently applied to screen the survival-related genes and hub genes, the intersection of which led to the identification of SERPINE1 that played key roles in the LGGs. The expression patterns, clinical features, and regulatory mechanisms of SERPINE1 in the LGGs were further analyzed by data mining of the TCGA database. What’s more, the above analyses of SERPINE1 were further validated in the LGG cohort from the CGGA database. Result We found that stromal and immune cell infiltrations were strongly related to the prognosis and malignancy of the LGGs. A total of 54 survival-related genes and 46 hub genes were screened out in the DEGs, within which SERPINE1 was identified to be significantly overexpressed in the LGG samples compared with the normal tissues. Moreover, the upregulation of SERPINE1 was more pronounced in the gliomas of WHO grade III and IDH wild type, and its expression was correlated with poor prognosis in the LGG patients. The independent prognostic value of SERPINE1 in the LGG patients was also confirmed by Cox regression analysis. In terms of the functions of SERPINE1, the results of enrichment analysis indicated that SERPINE1 was mainly enriched in the immune‐related biological processes and signaling pathways. Furthermore, it was closely associated with infiltrations of immune cells in the LGG microenvironment and acted synergistically with PD1, PD-L1, PD-L2. Conclusion These findings proved that SERPINE1 could serve as a prognostic biomarker and potential immunotherapy target of LGGs.
Collapse
Affiliation(s)
- Xiaoming Huang
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fenglin Zhang
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dong He
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoshuai Ji
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiajia Gao
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenqing Liu
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunda Wang
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qian Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Xin
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurosurgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, China.,Shandong Medicine and Health Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
40
|
Wang H, Guo Y, Gan S, Liu H, Chen Q, Yuan A, Hu Y, Wu J. Photosynthetic Microorganisms-Based Biophotothermal Therapy with Enhanced Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007734. [PMID: 33738929 DOI: 10.1002/smll.202007734] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/21/2021] [Indexed: 06/12/2023]
Abstract
The production of oxygen by photosynthetic microorganisms (PSMs) has recently attracted interest concerning the in vivo treatment of multiple diseases for their photosynthetic oxygen production in vivo, since PSMs have good biological safety. Here, the first evidence that PSMs can be used as a photothermal source to perform biophotothermal therapy (bio-PTT) is provided. In vitro and in vivo experiments proved that PSMs can generate heat for the direct elimination of tumors and release a series of pathogen-associated molecular patterns and adjuvants for immune stimulation under light irradiation. Bio-PTT enabled a local tumor inhibition rate exceeding 90% and an abscopal tumor inhibition rate exceeding 75%. This strategy also produced a stronger antitumor immune memory effect to prevent tumor recurrence. The bio-PTT strategy provides a novel direction for photothermal therapy as it simultaneously produces local and abscopal antitumor effects.
Collapse
Affiliation(s)
- Haoran Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Yunfei Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Shaoju Gan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Honghui Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
- Institute of Drug R&D, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
- Institute of Drug R&D, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
- Institute of Drug R&D, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
41
|
Li Z, Liu Y, Fang X, Shu Z. Nanomaterials Enhance the Immunomodulatory Effect of Molecular Targeted Therapy. Int J Nanomedicine 2021; 16:1631-1661. [PMID: 33688183 PMCID: PMC7935456 DOI: 10.2147/ijn.s290346] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/23/2021] [Indexed: 01/22/2023] Open
Abstract
Molecular targeted therapy, a tumor therapy strategy that inhibits specific oncogenic targets, has been shown to modulate the immune response. In addition to directly inhibiting the proliferation and metastasis of tumor cells, molecular targeted drugs can activate the immune system through a variety of mechanisms, including by promoting tumor antigen processing and presentation, increasing intratumoral T cell infiltration, enhancing T cell activation and function, and attenuating the immunosuppressive effect of the tumor microenvironment. However, poor water solubility, insufficient accumulation at the tumor site, and nonspecific targeting of immune cells limit their application. To this end, a variety of nanomaterials have been developed to overcome these obstacles and amplify the immunomodulatory effects of molecular targeted drugs. In this review, we summarize the impact of molecular targeted drugs on the antitumor immune response according to their mechanisms, highlight the advantages of nanomaterials in enhancing the immunomodulatory effect of molecular targeted therapy, and discuss the current challenges and future prospects.
Collapse
Affiliation(s)
- Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Yilun Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
42
|
Wang Y, Shen N, Wang Y, Zhang Y, Tang Z, Chen X. Self-Amplifying Nanotherapeutic Drugs Homing to Tumors in a Manner of Chain Reaction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2002094. [PMID: 33382144 DOI: 10.1002/adma.202002094] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/26/2020] [Indexed: 06/12/2023]
Abstract
Active tumor-targeting drug delivery has great potency in cancer therapy. However, the targeting efficiency of traditional active tumor-targeting nanotherapeutic drugs is limited by the scarcity of their accessible targets/receptors in tumors. Here, a novel self-amplifying tumor-targeting strategy with a chain reaction mechanism is developed. A coagulation targeting peptide (GNQEQVSPLTLLKXC, termed A15)-decorated poly(L-glutamic acid)-graft-maleimide poly(ethylene glycol)/combretastatin A4 conjugate (A15-PLG-CA4) is prepared to obtain a self-amplifying nanotherapeutic platform homing to tumors. After administration to tumor-bearing mice, A15-PLG-CA4 starts a chain reaction cycle consisting of intratumoral hemorrhage, target FXIIIa amplification, blood clot binding, and CA4 release in tumors. In this way, A15-PLG-CA4 increases the level of its accessible targets (FXIIIa) in a manner of chain reaction. The FXIIIa activity at 8 h is 4.1-fold more than the one at 0 h in the C26 tumors treated with A15-PLG-CA4. The total CA4 concentration at 24 h is 2.9-fold more than the control. A15-PLG-CA4 shows a significantly higher antitumor effect against large C26 tumors (≈500 mm3 ) thanks to the remarkable tumor-targeting ability compared with the control. Therefore, this report highlights the potential of the self-amplifying tumor-targeting strategy in the development of next generation active tumor-targeting nanotherapeutic drugs for tumor therapy.
Collapse
Affiliation(s)
- Yue Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Ying Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
43
|
Li C, Xu X, Wei S, Jiang P, Xue L, Wang J. Tumor-associated macrophages: potential therapeutic strategies and future prospects in cancer. J Immunother Cancer 2021; 9:jitc-2020-001341. [PMID: 33504575 PMCID: PMC8728363 DOI: 10.1136/jitc-2020-001341] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2020] [Indexed: 12/11/2022] Open
Abstract
Macrophages are the most important phagocytes in vivo. However, the tumor microenvironment can affect the function and polarization of macrophages and form tumor-associated macrophages (TAMs). Usually, the abundance of TAMs in tumors is closely associated with poor prognosis. Preclinical studies have identified important pathways regulating the infiltration and polarization of TAMs during tumor progression. Furthermore, potential therapeutic strategies targeting TAMs in tumors have been studied, including inhibition of macrophage recruitment to tumors, functional repolarization of TAMs toward an antitumor phenotype, and other therapeutic strategies that elicit macrophage-mediated extracellular phagocytosis and intracellular destruction of cancer cells. Therefore, with the increasing impact of tumor immunotherapy, new antitumor strategies to target TAMs are now being discussed.
Collapse
Affiliation(s)
- Chunxiao Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Xiaofei Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Department of Obstetrics and Gynecology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Shuhua Wei
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Ping Jiang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Lixiang Xue
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | | | | |
Collapse
|
44
|
Zhao D, Huang X, Zhang Z, Ding J, Cui Y, Chen X. Engineered nanomedicines for tumor vasculature blockade therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1691. [PMID: 33480163 DOI: 10.1002/wnan.1691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022]
Abstract
Tumor vasculature blockade therapy (TVBT), including angiogenesis inhibition, vascular disruption, and vascular infarction, provides a promising treatment modality for solid tumors. However, low selectivity, drug resistance, and possible severe side effects have limited the clinical transformation of TVBT. Engineered nanoparticles offer potential solutions, including prolonged circulation time, targeted transportation, and controlled release of TVBT agents. Moreover, engineered nanomedicines provide a promising combination platform of TVBT with chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, ultrasound therapy, and gene therapy. In this article, we offer a comprehensive summary of the current progress of engineered nanomedicines for TVBT and also discuss current deficiencies and future directions for TVBT development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Xu Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
45
|
Robust and smart polypeptide-based nanomedicines for targeted tumor therapy. Adv Drug Deliv Rev 2020; 160:199-211. [PMID: 33137364 DOI: 10.1016/j.addr.2020.10.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Nanomedicines based on synthetic polypeptides are among the most versatile and advanced platforms for tumor therapy. Notably, several polypeptide-based nanodrugs are currently under human clinical assessments. The previous (pre)clinical studies clearly show that dynamic stability (i.e. stable in circulation while destabilized in tumor) of nanomedicines plays a vital role in their anti-tumor performance. Various strategies have recently been developed to design dynamically stabilized polypeptide-based nanomedicines by e.g. crosslinking the nanovehicles with acid, reactive oxygen species (ROS), glutathione (GSH), or photo-sensitive linkers, inter-crosslinking between vehicles and drugs, introducing π-π stacking or lipid-lipid interactions in the nanovehicles, chemically conjugating drugs to vehicles, and forming unimolecular micelles. Interestingly, these robust and smart nanodrugs have demonstrated improved tumor targetability, anti-tumor efficacy, as well as safety profiles in different tumor models. In this review, representative strategies to robust and smart polypeptide-based nanomedicines for targeted treatment of varying malignancies are highlighted. The exciting development of dynamic nanomedicines will foresee further increasing clinical translation in the future.
Collapse
|
46
|
Fan D, Zeng C, Wang S, Han J, Zhu L, Zhao H, Zhang Y, Lu J, Xu Y. Forkhead box P3 promotes breast cancer cell apoptosis by regulating programmed cell death 4 expression. Oncol Lett 2020; 20:292. [PMID: 33101486 PMCID: PMC7576988 DOI: 10.3892/ol.2020.12155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
Forkhead box P3 (FOXP3), an X-linked tumor suppressor gene, plays an important role in breast cancer. However, the biological functions of FOXP3 in breast cancer apoptosis remain unclear. To investigate the underlying genes and networks regulated by FOXP3 in breast cancer, RNA sequencing was performed to compare FOXP3-overexpressing MDA-MB-231 cells and control MDA-MB-231 cells. Differentially expressed genes were identified, and functional enrichment analysis comparing the two groups was performed. The differentially expressed genes were mainly enriched in phagosomes, oxytocin, serotonergic synapses and the phospholipase D signaling pathway. Furthermore, gene set enrichment analysis revealed the enrichment of a gene signature associated with apoptosis in FOXP3-overexpressing MDA-MB-231 cells compared with wild-type cells. Further analysis showed that programmed cell death 4 (PDCD4), a key molecule involved in apoptosis, was overexpressed in FOXP3-MDA-MB-231 cells. Reverse transcription-quantitative PCR and western blotting showed that FOXP3 upregulated the expression of PDCD4 in breast cancer cells. Clinical sample analysis using a public database showed that the expression level of PDCD4 was associated with breast cancer clinical stages. Overall, the present study suggested that FOXP3 can promote the apoptosis of breast cancer cells by upregulating the expression of PDCD4, thus exerting a tumor suppressive function.
Collapse
Affiliation(s)
- Dong Fan
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China.,Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Cheng Zeng
- Department of Chinese Materia Medical and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China.,Department of Pharmacy, Central Theater Command Air Force Hospital, Datong, Shaanxi 037000, P.R. China
| | - Shuming Wang
- Outpatient Department of Rocket Engineering University, The 986th Hospital of Air Force, Air Force Medical University, Xi'an, Shaanxi 710054, P.R. China
| | - Jun Han
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Liaoliao Zhu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jianguo Lu
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Ying Xu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of The Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
47
|
Yu H, Bao Y, Xu C, Chen L, Tang Z. Poly(L-Glutamic Acid)-Drug Conjugates for Chemo- and Photodynamic Combination Therapy. Macromol Biosci 2020; 21:e2000192. [PMID: 33043592 DOI: 10.1002/mabi.202000192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/26/2020] [Indexed: 11/09/2022]
Abstract
Despite the polymeric vascular disrupting agent (poly(L -glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4) nanoparticles can efficiently inhibit cancer growth, their further application is still a challenge owing to the tumor recurrence and metastasis after treatment. In this study, two poly(L -glutamic acid)-drug conjugates for chemo-and photodynamic combination therapy are fabricated. PLG-g-mPEG-CA4 nanoparticles are prepared by combretastatin A4 (CA4) and poly(L -glutamic acid)-graft-methoxy poly(ethylene glycol) (PLG-g-mPEG) using the Yamaguchi esterification reaction. PLG-g-mPEG-TPP (TPP: 5, 10, 15, 20-tetraphenylporphyrin) nanoparticles are constructed using PLG-g-mPEG and amine porphyrin through condensation reaction between carboxyl group of PLG-g-mPEG and amino group of porphyrin. The results showed that PLG-g-mPEG-CA4 nanoparticles have good antitumor ability. PLG-g-mPEG-TPP nanoparticles can produce singlet oxygen under the laser irradiation. Moreover, the combined therapy of PLG-g-mPEG-CA4 and PLG-g-mPEG-TPP nanoparticles has higher antitumor effect than the single chemotherapy or the single photodynamic therapy in vitro. The combination of CA4 nondrug and photodynamic therapy provides a new insight for enhancing the tumor therapeutic effect with vascular disrupting agents and other therapy.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Yanli Bao
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China
| | - Caina Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Li Chen
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| |
Collapse
|
48
|
A 2-step synthesis of Combretastatin A-4 and derivatives as potent tubulin assembly inhibitors. Bioorg Med Chem 2020; 28:115684. [DOI: 10.1016/j.bmc.2020.115684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 11/19/2022]
|
49
|
Pecyna P, Wargula J, Murias M, Kucinska M. More Than Resveratrol: New Insights into Stilbene-Based Compounds. Biomolecules 2020; 10:E1111. [PMID: 32726968 PMCID: PMC7465418 DOI: 10.3390/biom10081111] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The concept of a scaffold concerns many aspects at different steps on the drug development path. In medicinal chemistry, the choice of relevant "drug-likeness" scaffold is a starting point for the design of the structure dedicated to specific molecular targets. For many years, the chemical uniqueness of the stilbene structure has inspired scientists from different fields such as chemistry, biology, pharmacy, and medicine. In this review, we present the outstanding potential of the stilbene-based derivatives. Naturally occurring stilbenes, together with powerful synthetic chemistry possibilities, may offer an excellent approach for discovering new structures and identifying their therapeutic targets. With the development of scientific tools, sophisticated equipment, and a better understanding of the disease pathogenesis at the molecular level, the stilbene scaffold has moved innovation in science. This paper mainly focuses on the stilbene-based compounds beyond resveratrol, which are particularly attractive due to their biological activity. Given the "fresh outlook" about different stilbene-based compounds starting from stilbenoids with particular regard to isorhapontigenin and methoxy- and hydroxyl- analogues, the update about the combretastatins, and the very often overlooked and underestimated benzanilide analogues, we present a new story about this remarkable structure.
Collapse
Affiliation(s)
- Paulina Pecyna
- Department of Genetics and Pharmaceutical Microbiology, University of Medical Sciences, Swiecickiego 4 Street, 60-781 Poznan, Poland;
| | - Joanna Wargula
- Department of Organic Chemistry, University of Medical Sciences, Grunwaldzka 6 Street, 60-780 Poznan, Poland;
| | - Marek Murias
- Department of Toxicology, University of Medical Sciences, Dojazd 30 Street, 60-631 Poznan, Poland;
| | - Malgorzata Kucinska
- Department of Toxicology, University of Medical Sciences, Dojazd 30 Street, 60-631 Poznan, Poland;
| |
Collapse
|
50
|
Abstract
The concept of a scaffold concerns many aspects at different steps on the drug development path. In medicinal chemistry, the choice of relevant "drug-likeness" scaffold is a starting point for the design of the structure dedicated to specific molecular targets. For many years, the chemical uniqueness of the stilbene structure has inspired scientists from different fields such as chemistry, biology, pharmacy, and medicine. In this review, we present the outstanding potential of the stilbene-based derivatives. Naturally occurring stilbenes, together with powerful synthetic chemistry possibilities, may offer an excellent approach for discovering new structures and identifying their therapeutic targets. With the development of scientific tools, sophisticated equipment, and a better understanding of the disease pathogenesis at the molecular level, the stilbene scaffold has moved innovation in science. This paper mainly focuses on the stilbene-based compounds beyond resveratrol, which are particularly attractive due to their biological activity. Given the "fresh outlook" about different stilbene-based compounds starting from stilbenoids with particular regard to isorhapontigenin and methoxy- and hydroxyl- analogues, the update about the combretastatins, and the very often overlooked and underestimated benzanilide analogues, we present a new story about this remarkable structure.
Collapse
|