1
|
Scampa M, Martineau J, Boet S, Pignel R, Kalbermatten DF, Oranges CM. Hyperbaric oxygen therapy outcomes in post-irradiated patient undergoing microvascular breast reconstruction: A preliminary retrospective comparative study. JPRAS Open 2024; 42:1-9. [PMID: 39279849 PMCID: PMC11399798 DOI: 10.1016/j.jpra.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/28/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Radiotherapy is a challenge in autologous breast reconstruction because of its impact on cutaneous and vascular systems. Hyperbaric oxygen therapy (HBOT) is a recognized treatment of radiation-related complications. We aimed to assess the impact of perioperative HBOT on irradiated breast microvascular reconstructive outcomes. Method We reviewed the medical charts of patients who received radiotherapy and then underwent secondary free autologous breast reconstruction at our institution. Data on demographics, HBOT protocol, intervention characteristics and post-operative complications were collected. Outcomes of the irradiated patients were then compared between the HBOT and non-HBOT groups. Results Fourteen patients were included (11 unilateral and 2 bilateral deep inferior epigastric artery perforator flaps and 1 free transverse rectus abdominis muscle flap). Seven patients received HBOT and 7 did not. In the non-HBOT group, there were 1 Clavien-Dindo grade II, 1 Clavien-Dindo grade IIIa and 2 Clavien-Dindo grade IIIb post-operative complications. In the HBOT group, there were 3 Clavien-Dindo grade I, 1 Clavien-Dindo grade IIIa and 2 Clavien-Dindo grade IIIb post-operative complications. The mean operative time was 452.3 minutes (SD ±62.4 minutes) for unilateral cases without HBOT and 457.8 minutes (SD ±102.1 minutes) with HBOT (p=0.913). Mean ischaemia time per flap without HBOT was 109.4 minutes (SD ±51.8 minutes) versus 80.1 minutes (SD ±37.7 minutes) in the HBOT group (p=0.249). Conclusion This study provides insights into the potential of HBOT treatment in preparing patients with irradiated breast cancer for secondary autologous reconstruction.
Collapse
Affiliation(s)
- Matteo Scampa
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Geneva University Hospitals, Geneva University, 1205, Geneva, Switzerland
| | - Jérôme Martineau
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Geneva University Hospitals, Geneva University, 1205, Geneva, Switzerland
| | - Sylvain Boet
- Subaquatic and Hyperbaric Medicine Unit, Division of Emergency Medicine, Department of Anesthesiology, Clinical Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, 1205, Geneva, Switzerland
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
- Ottawa Hospital Research Institute, Clinical Epidemiology Program, Department of Innovation in Medical Education, University of Ottawa, Ottawa, ON, K1H 8L6, Canada
- Institut du Savoir Montfort, Ottawa, ON, K1K 0T2, Canada
| | - Rodrigue Pignel
- Subaquatic and Hyperbaric Medicine Unit, Division of Emergency Medicine, Department of Anesthesiology, Clinical Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, 1205, Geneva, Switzerland
| | - Daniel F Kalbermatten
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Geneva University Hospitals, Geneva University, 1205, Geneva, Switzerland
| | - Carlo M Oranges
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Geneva University Hospitals, Geneva University, 1205, Geneva, Switzerland
| |
Collapse
|
2
|
Scarmelotto A, Delprat V, Michiels C, Lucas S, Heuskin AC. The oxygen puzzle in FLASH radiotherapy: A comprehensive review and experimental outlook. Clin Transl Radiat Oncol 2024; 49:100860. [PMID: 39381632 PMCID: PMC11458961 DOI: 10.1016/j.ctro.2024.100860] [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: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
FLASH radiotherapy is attracting increasing interest because it maintains tumor control while inflicting less damage to normal tissues compared to conventional radiotherapy. This sparing effect, the so-called FLASH effect, is achieved when radiation is delivered at ultra-high dose rates (≥40 Gy/s). Although the FLASH effect has already been demonstrated in several preclinical models, a complete mechanistic description explaining why tumors and normal tissues respond differently is still missing. None of the current hypotheses fully explains the experimental evidence. A common point between many of these is the role of oxygen, which is described as a major factor, either through transient hypoxia in the form of dissolved molecules, or reactive oxygen species (ROS). Therefore, this review focuses on both forms of this molecule, retracing old and more recent theories, while proposing new mechanisms that could provide a complete description of the FLASH effect based on preclinical and experimental evidence. In addition, this manuscript describes a set of experiments designed to provide the FLASH community with new tools for exploring the post-irradiation fate of ROS and their potential biological implications.
Collapse
Affiliation(s)
- Andrea Scarmelotto
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Victor Delprat
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute For Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Stéphane Lucas
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
- Ion Beam Application (IBA), Chemin du Cyclotron, 6, B-1348 Louvain-La-Neuve, Belgium
| | - Anne-Catherine Heuskin
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| |
Collapse
|
3
|
Rafiyan M, Tootoonchi E, Golpour M, Davoodvandi A, Reiter RJ, Asemi R, Sharifi M, Rasooli Manesh SM, Asemi Z. Melatonin for gastric cancer treatment: where do we stand? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03451-7. [PMID: 39287677 DOI: 10.1007/s00210-024-03451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Gastric cancer (GC) is the third leading reason of death in men and the fourth in women. Studies have documented an inhibitory function of melatonin on the proliferation, progression and invasion of GC cells. MicroRNAs (miRNAs) are small, non-coding RNAs that play an important function in regulation of biological processes and gene expression of the cells. Some studies reported that melatonin can suppress the progression of GC by regulating the exosomal miRNAs. Thus, melatonin represents a promising potential therapeutic agent for subjects with GC. Herein, we evaluate the existing data of both in vivo and in vitro studies to clarify the molecular processes involved in the therapeutic effects of melatonin in GC. The data emphasize the critical function of melatonin in several signaling ways by which it may inhibit cancer cell proliferation, decrease chemo-resistance, induce apoptosis as well as limit invasion, angiogenesis, and metastasis. This review provides a resource that identifies some of the mechanisms by which melatonin controls GC enlargement. In light of the findings, melatonin should be considered a novel and testable therapeutic mediator for GC treatment.
Collapse
Affiliation(s)
- Mahdi Rafiyan
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Elham Tootoonchi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahdieh Golpour
- Student Research Committee, Mazandarn University of Medical Sciences, Sari, Mazandaran, Iran
| | - Amirhossein Davoodvandi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Ma K, Wang S, Ma Y, Zeng L, Xu K, Mu N, Lai Y, Shi Y, Yang C, Chen B, Quan Y, Li L, Lu Y, Yang Y, Liu Y, Hu R, Wang X, Chen Y, Bian X, Feng H, Li F, Chen T. Increased oxygen stimulation promotes chemoresistance and phenotype shifting through PLCB1 in gliomas. Drug Resist Updat 2024; 76:101113. [PMID: 39053384 DOI: 10.1016/j.drup.2024.101113] [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: 01/22/2024] [Revised: 04/24/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
Gliomas, the most common CNS (central nerve system) tumors, face poor survival due to severe chemoresistance exacerbated by hypoxia. However, studies on whether altered hypoxic conditions benefit for chemo-sensitivity and how gliomas react to increased oxygen stimulation are limited. In this study, we demonstrated that increased oxygen stimulation promotes glioma growth and chemoresistance. Mechanically, increased oxygen stimulation upregulates miR-1290 levels. miR-1290, in turn, downregulates PLCB1, while PLCB1 facilitates the proteasomal degradation of β-catenin and active-β-catenin by increasing the proportion of ubiquitinated β-catenin in a destruction complex-independent mechanism. This process inhibits PLCB1 expression, leads to the accumulation of active-β-catenin, boosting Wnt signaling through an independent mechanism and ultimately promoting chemoresistance in glioma cells. Pharmacological inhibition of Wnt by WNT974 could partially inhibit glioma volume growth and prolong the shortened survival caused by increased oxygen stimulation in a glioma-bearing mouse model. Moreover, PLCB1, a key molecule regulated by increased oxygen stimulation, shows promising predictive power in survival analysis and has great potential to be a biomarker for grading and prognosis in glioma patients. These results provide preliminary insights into clinical scenarios associated with altered hypoxic conditions in gliomas, and introduce a novel perspective on the role of the hypoxic microenvironment in glioma progression. Furthermore, the outcomes reveal the potential risks of utilizing hyperbaric oxygen treatment (HBOT) in glioma patients, particularly when considering HBOT as a standalone option to ameliorate neuro-dysfunctions or when combining HBOT with a single chemotherapy agent without radiotherapy.
Collapse
Affiliation(s)
- Kang Ma
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi Wang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yingjie Ma
- Medical Data Science Academy, Chongqing Medical University, Chongqing, China
| | - Lan Zeng
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Kai Xu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ning Mu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Lai
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yaning Shi
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuanyan Yang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Beike Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yulian Quan
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Li
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongling Lu
- Medical Research Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yan Liu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Rong Hu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoming Wang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yujie Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Hua Feng
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Fei Li
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Tunan Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
5
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self-Catalytic NO/O 2 Gas-Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403921. [PMID: 39101290 DOI: 10.1002/adma.202403921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Radiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor-targeted "prosthetic-Arginine" coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO-donating Fmoc-protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu-HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2-transporting ability of heme, HRRu-HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu-HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu-HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment, Henan, 450001, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
6
|
Alfei S, Schito GC, Schito AM, Zuccari G. Reactive Oxygen Species (ROS)-Mediated Antibacterial Oxidative Therapies: Available Methods to Generate ROS and a Novel Option Proposal. Int J Mol Sci 2024; 25:7182. [PMID: 39000290 PMCID: PMC11241369 DOI: 10.3390/ijms25137182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The increasing emergence of multidrug-resistant (MDR) pathogens causes difficult-to-treat infections with long-term hospitalizations and a high incidence of death, thus representing a global public health problem. To manage MDR bacteria bugs, new antimicrobial strategies are necessary, and their introduction in practice is a daily challenge for scientists in the field. An extensively studied approach to treating MDR infections consists of inducing high levels of reactive oxygen species (ROS) by several methods. Although further clinical investigations are mandatory on the possible toxic effects of ROS on mammalian cells, clinical evaluations are extremely promising, and their topical use to treat infected wounds and ulcers, also in presence of biofilm, is already clinically approved. Biochar (BC) is a carbonaceous material obtained by pyrolysis of different vegetable and animal biomass feedstocks at 200-1000 °C in the limited presence of O2. Recently, it has been demonstrated that BC's capability of removing organic and inorganic xenobiotics is mainly due to the presence of persistent free radicals (PFRs), which can activate oxygen, H2O2, or persulfate in the presence or absence of transition metals by electron transfer, thus generating ROS, which in turn degrade pollutants by advanced oxidation processes (AOPs). In this context, the antibacterial effects of BC-containing PFRs have been demonstrated by some authors against Escherichia coli and Staphylococcus aureus, thus giving birth to our idea of the possible use of BC-derived PFRs as a novel method capable of inducing ROS generation for antimicrobial oxidative therapy. Here, the general aspects concerning ROS physiological and pathological production and regulation and the mechanism by which they could exert antimicrobial effects have been reviewed. The methods currently adopted to induce ROS production for antimicrobial oxidative therapy have been discussed. Finally, for the first time, BC-related PFRs have been proposed as a new source of ROS for antimicrobial therapy via AOPs.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano, 4, 16148 Genoa, Italy
| | - Gian Carlo Schito
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
| | - Anna Maria Schito
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
| | - Guendalina Zuccari
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano, 4, 16148 Genoa, Italy
| |
Collapse
|
7
|
Xu KF, Wu SY, Wang Z, Guo Y, Zhu YX, Li C, Shan BH, Zhang X, Liu X, Wu FG. Hyperbaric oxygen enhances tumor penetration and accumulation of engineered bacteria for synergistic photothermal immunotherapy. Nat Commun 2024; 15:5147. [PMID: 38886343 PMCID: PMC11183253 DOI: 10.1038/s41467-024-49156-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/25/2024] [Indexed: 06/20/2024] Open
Abstract
Bacteria-mediated cancer therapeutic strategies have attracted increasing interest due to their intrinsic tumor tropism. However, bacteria-based drugs face several challenges including the large size of bacteria and dense extracellular matrix, limiting their intratumoral delivery efficiency. In this study, we find that hyperbaric oxygen (HBO), a noninvasive therapeutic method, can effectively deplete the dense extracellular matrix and thus enhance the bacterial accumulation within tumors. Inspired by this finding, we modify Escherichia coli Nissle 1917 (EcN) with cypate molecules to yield EcN-cypate for photothermal therapy, which can subsequently induce immunogenic cell death (ICD). Importantly, HBO treatment significantly increases the intratumoral accumulation of EcN-cypate and facilitates the intratumoral infiltration of immune cells to realize desirable tumor eradication through photothermal therapy and ICD-induced immunotherapy. Our work provides a facile and noninvasive strategy to enhance the intratumoral delivery efficiency of natural/engineered bacteria, and may promote the clinical translation of bacteria-mediated synergistic cancer therapy.
Collapse
Affiliation(s)
- Ke-Fei Xu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Shun-Yu Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Zihao Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Yuxin Guo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Ya-Xuan Zhu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Chengcheng Li
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Bai-Hui Shan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Xinping Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China.
| |
Collapse
|
8
|
Hajikarimloo B, Kavousi S, Jahromi GG, Mehmandoost M, Oraee-Yazdani S, Fahim F. Hyperbaric Oxygen Therapy as an Alternative Therapeutic Option for Radiation-Induced Necrosis Following Radiotherapy for Intracranial Pathologies. World Neurosurg 2024; 186:51-61. [PMID: 38325705 DOI: 10.1016/j.wneu.2024.01.161] [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: 01/13/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Radiotherapy (RT) is a feasible adjuvant therapeutic option for managing intracranial pathologies. One of the late complications of RT that frequently develops within months following RT is radiation necrosis (RN). Corticosteroids are the first-line therapeutic option for RNs; however, in case of unfavorable outcomes or intolerability, several other options, including bevacizumab, laser interstitial thermal therapy, surgery, and hyperbaric oxygen therapy (HBOT). Our goal was to investigate the feasibility and efficacy of the application of HBOT in RNs following RT and help physicians make decisions based on the latest data in the literature. METHODS We provide a comprehensive review of the literature on the current issues of utilization of HBOT in RNs. RESULTS We included 11 studies with a total of 46 patients who underwent HBOT. Most of the cases were diagnosed with brain tumors or arteriovenous malformations. Improvement was achieved in most of the cases. DISCUSSION HBOT is a noninvasive therapeutic intervention that can play a role in adjuvant therapy concurrent with RT and chemotherapy and treating RNs. HBOT resolves the RN through 3 mechanisms, including angiogenesis, anti-inflammatory modulation, and cellular repair. Previous studies demonstrated that HBOT is a feasible and well-tolerated therapeutic option that has shown promising results in improving clinical and radiological outcomes in intracranial RNs. Complications of HBOT are usually mild and reversible. CONCLUSIONS HBOT is a feasible and effective therapeutic option in steroid-refractory RNs and is associated with favorable outcomes and a low rate of side effects.
Collapse
Affiliation(s)
- Bardia Hajikarimloo
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Shohada Tajrish Hospital, Tehran, Iran
| | - Shahin Kavousi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Ghaffaripour Jahromi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Mehmandoost
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Shohada Tajrish Hospital, Tehran, Iran
| | - Farzan Fahim
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Shohada Tajrish Hospital, Tehran, Iran.
| |
Collapse
|
9
|
Epel B, Viswakarma N, Sundramoorthy SV, Pawar NJ, Kotecha M. Oxygen Imaging of a Rabbit Tumor Using a Human-Sized Pulse Electron Paramagnetic Resonance Imager. Mol Imaging Biol 2024; 26:403-410. [PMID: 37715089 DOI: 10.1007/s11307-023-01852-3] [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: 05/08/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE Spatial heterogeneity in tumor hypoxia is one of the most important factors regulating tumor growth, development, aggressiveness, metastasis, and affecting treatment outcome. Most solid tumors are known to have hypoxia or low oxygen levels (pO2 ≤10 torr). Electron paramagnetic resonance oxygen imaging (EPROI) is an emerging oxygen mapping technology. EPROI utilizes the linear relationship between the relaxation rates of the injectable OX071 trityl spin probe and the partial oxygen pressure (pO2). However, most of the EPROI studies have been limited to mouse models of solid tumors because of the instrument-size limitations. The purpose of this work was to develop a human-sized 9-mT (250 MHz resonance frequency, 60 cm bore size) pulse EPROI instrument and evaluate its performance with rabbit VX-2 tumor oxygen imaging. METHODS A New Zealand white rabbit with a 3.2-cm VX-2 tumor in the calf muscle was imaged using the human-sized EPROI instrument and a 2.25-in. ID volume coil. The animal received a ~8-min intravenous injection of OX071 (5.2 mL total volume at 72 mM concentration) and, after 75 min, an intratumoral injection (120 μL total at 5 mM OX071 concentration) and underwent EPROI. At the end of the experiments, MRI was performed using a preclinical 9.4-T MRI system to outline the tumor boundaries. RESULTS For the first time, a human-sized pulse EPROI instrument with a 60-cm bore size/250-MHz frequency was built and evaluated using rabbit tumor oxygen imaging. For the first time, the systemic IV injection of the oxygen-sensitive trityl OX071 spin probe was used for an animal of this size. The resulting EPROI image from the IV injection showed complete tumor coverage. The image obtained after intratumoral injection showed localized coverage in the upper lobe of the tumor, demonstrating the need for improved intratumoral injection protocol. CONCLUSIONS This study demonstrates the performance of the world's first human-sized pulse EPROI instrument. It also demonstrates that the EPROI of larger animals can be performed using the systemic injection of a manageable amount of the spin probe. This brings EPROI one step closer to clinical applications in cancer therapies. Oxygen imaging is a platform technology, and the instrument and techniques developed here will also be useful for other clinical applications.
Collapse
Affiliation(s)
- Boris Epel
- O2M Technologies, LLC, Chicago, IL, 60612, USA.
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, 60637, USA.
| | | | | | | | | |
Collapse
|
10
|
Ding Y, Luan W, Wang Z, Xie B, Sun C. HBO regulates the Warburg effect of hypoxic HCC cells through miR-103a-3p/TRIM35. Discov Oncol 2024; 15:125. [PMID: 38642184 PMCID: PMC11032302 DOI: 10.1007/s12672-024-00985-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/18/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND There are a lot of studies on the treatment of tumors with hyperbaric oxygen, while most of them are in breast cancer, prostate cancer and so on. However, there are still few studies on hyperbaric oxygen in treating hepatocellular carcinoma (HCC). According to the current data, hyperbaric oxygen is an effective means to intervene in tumors. The Warburg effect is a unique marker of glucose metabolism in tumors related to hypoxia, making it possible for hyperbaric oxygen to interfere with the tumor through the Warburg effect. METHOD We used the hypoxia/hyperbaric oxygen(HBO)-exposed HCC cells for in vitro studies. Glucose uptake, lactic acid, and adenosine triphosphate (ATP) assessed the Warburg effect. The expression of miR-103a-3p in HCC was detected by using qRT-PCR. The effect of miR-103a-3p/TRIM35 expression level on the cells was measured using the CCK8 method and flow cytometry. The molecular biological mechanism of miR-103a-3p in HCC was examined using the luciferase reporter, MS2-RIP assays. RESULT HBO inhibited the Warburg effect in hypoxic HCC cells. HBO suppressed the expression of miR-103a-3p in hypoxic HCC cells, and miR-103a-3p inhibited the expression of TRIM35 in hypoxic HCC cells. With HBO exposure, miR-103a-3p/TRIM35 regulated the Warburg effect of hypoxic HCC cells. CONCLUSION These findings reveal that HBO regulates the Warburg effect of hypoxic HCC cells through miR-103a-3p/TRIM35 and inhibits tumor growth.
Collapse
Affiliation(s)
- Yuting Ding
- Department of Rehabilitation, Changshu No. 2 People's Hospital (Changshu Hospital affiliated the Nantong University), Changshu, 215500, Jiangsu, China.
| | - Wenkang Luan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Wang
- School of Medicine, JiangSu University, Zhenjiang, 212013, Jiangsu, China
| | - Bo Xie
- Department of Rehabilitation, Changshu No. 2 People's Hospital (Changshu Hospital affiliated the Nantong University), Changshu, 215500, Jiangsu, China
| | - Chengfa Sun
- Department of Neurosurgery, Changshu No. 2 People's Hospital (Changshu Hospital affiliated the Nantong University), Changshu, 215500, Jiangsu, China
| |
Collapse
|
11
|
Martinez P, Baghli I, Gourjon G, Seyfried TN. Mitochondrial-Stem Cell Connection: Providing Additional Explanations for Understanding Cancer. Metabolites 2024; 14:229. [PMID: 38668357 PMCID: PMC11051897 DOI: 10.3390/metabo14040229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The cancer paradigm is generally based on the somatic mutation model, asserting that cancer is a disease of genetic origin. The mitochondrial-stem cell connection (MSCC) proposes that tumorigenesis may result from an alteration of the mitochondria, specifically a chronic oxidative phosphorylation (OxPhos) insufficiency in stem cells, which forms cancer stem cells (CSCs) and leads to malignancy. Reviewed evidence suggests that the MSCC could provide a comprehensive understanding of all the different stages of cancer. The metabolism of cancer cells is altered (OxPhos insufficiency) and must be compensated by using the glycolysis and the glutaminolysis pathways, which are essential to their growth. The altered mitochondria regulate the tumor microenvironment, which is also necessary for cancer evolution. Therefore, the MSCC could help improve our understanding of tumorigenesis, metastases, the efficiency of standard treatments, and relapses.
Collapse
Affiliation(s)
- Pierrick Martinez
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 84140 Montfavet, France;
| | - Ilyes Baghli
- International Society for Orthomolecular Medicine, Toronto, ON M4B 3M9, Canada;
| | - Géraud Gourjon
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 84140 Montfavet, France;
| | | |
Collapse
|
12
|
Liu B, Du F, Feng Z, Xiang X, Guo R, Ma L, Zhu B, Qiu L. Ultrasound-augmented cancer immunotherapy. J Mater Chem B 2024; 12:3636-3658. [PMID: 38529593 DOI: 10.1039/d3tb02705h] [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: 03/27/2024]
Abstract
Cancer is a growing worldwide health problem with the most broadly studied treatments, in which immunotherapy has made notable advancements in recent years. However, innumerable patients have presented a poor response to immunotherapy and simultaneously experienced immune-related adverse events, with failed therapeutic results and increased mortality rates. Consequently, it is crucial to develop alternate tactics to boost therapeutic effects without producing negative side effects. Ultrasound is considered to possess significant therapeutic potential in the antitumor field because of its inherent characteristics, including cavitation, pyrolysis, and sonoporation. Herein, this timely review presents the comprehensive and systematic research progress of ultrasound-enhanced cancer immunotherapy, focusing on the various ultrasound-related mechanisms and strategies. Moreover, this review summarizes the design and application of current sonosensitizers based on sonodynamic therapy, with an attempt to provide guidance on new directions for future cancer therapy.
Collapse
Affiliation(s)
- Bingjie Liu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Fangxue Du
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ziyan Feng
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xi Xiang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruiqian Guo
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bihui Zhu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Fijardo M, Kwan JYY, Bissey PA, Citrin DE, Yip KW, Liu FF. The clinical manifestations and molecular pathogenesis of radiation fibrosis. EBioMedicine 2024; 103:105089. [PMID: 38579363 PMCID: PMC11002813 DOI: 10.1016/j.ebiom.2024.105089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 04/07/2024] Open
Abstract
Advances in radiation techniques have enabled the precise delivery of higher doses of radiotherapy to tumours, while sparing surrounding healthy tissues. Consequently, the incidence of radiation toxicities has declined, and will likely continue to improve as radiotherapy further evolves. Nonetheless, ionizing radiation elicits tissue-specific toxicities that gradually develop into radiation-induced fibrosis, a common long-term side-effect of radiotherapy. Radiation fibrosis is characterized by an aberrant wound repair process, which promotes the deposition of extensive scar tissue, clinically manifesting as a loss of elasticity, tissue thickening, and organ-specific functional consequences. In addition to improving the existing technologies and guidelines directing the administration of radiotherapy, understanding the pathogenesis underlying radiation fibrosis is essential for the success of cancer treatments. This review integrates the principles for radiotherapy dosimetry to minimize off-target effects, the tissue-specific clinical manifestations, the key cellular and molecular drivers of radiation fibrosis, and emerging therapeutic opportunities for both prevention and treatment.
Collapse
Affiliation(s)
- Mackenzie Fijardo
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Yin Yee Kwan
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States of America
| | - Kenneth W Yip
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Fei-Fei Liu
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Zuo YC, Huo CM, Chen Y, Ding PL, Tong SY, Xue W, Zhu JY. Cancer-Thylakoid Hybrid Membrane Camouflaged Thulium Oxide Nanoparticles with Oxygen Self-Supply Capability for Tumor-Homing Phototherapy. Adv Healthc Mater 2024; 13:e2303779. [PMID: 38288884 DOI: 10.1002/adhm.202303779] [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: 10/31/2023] [Revised: 01/06/2024] [Indexed: 02/13/2024]
Abstract
Nanomaterials that generate reactive oxygen species (ROS) upon light irradiation have significant applications in various fields, including photodynamic therapy (PDT) that is widely recognized as a highly momentous strategy for the eradication of cancer cells. However, the ROS production rate of photosensitizers, as well as the tumor hypoxia environment, are two major challenges that restrict the widespread application of PDT. In this study, a cancer-thylakoid hybrid membrane-camouflaged thulium oxide nanoparticles (Tm2O3) for tumor-homing phototherapy through dual-stage-light-guided ROS generation and oxygen self-supply is developed. Tm2O3 as a type II photosensitizer are viable for NIR-stimulated ROS generation due to the unique energy levels, large absorption cross section, and long lifetime of the 3H4 state of Tm ions. The thylakoid membrane (TK) plays a catalase-like role in converting hydrogen peroxide into oxygen and also acts as a natural photosensitizer that can generate lethal ROS through electron transfer when exposed to light. In addition, fluorescence dye DiR is embedded in the hybrid membrane for in vivo tracing as well as photothermal therapy. Results show that tumors in Tm2O3@TK-M/DiR group are effectively ablated following dual-stage-light irradiation, highlighting the promising potential of rare-earth element-based type II photosensitizers in various applications.
Collapse
Affiliation(s)
- Yu-Cheng Zuo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Cong-Min Huo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yu Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Peng-Li Ding
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Si-Ye Tong
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Jing-Yi Zhu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
15
|
Liu J, An W, Zhao Q, Liu Z, Jiang Y, Li H, Wang D. Hyperbaric oxygen enhances X-ray induced ferroptosis in oral squamous cell carcinoma cells. Oral Dis 2024; 30:116-127. [PMID: 36495316 DOI: 10.1111/odi.14461] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 10/10/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The objective of this study was to investigate the combined effect of X-ray radiation (IR) and hyperbaric oxygen (HBO) on oral squamous cell carcinoma (OSCC) cells and to explore the possible molecular mechanism. METHODS The OSCC cells were treated with or without IR, together with or without HBO co-exposure. Cells were transfected with specific plasmids using Lipofectamine 2000. The cell varieties, apoptosis markers, and ferroptosis markers were determined by using appropriate method. OSCC xenograft mice model was categorized into several subgroups according to the specific treatement. GPX4 expressions were determined by immunohistochemistry (IHC) in OSCC tissues and were tested by ELISA in serums from OSCC patients. RESULTS The co-exposure of IR and HBO significantly strengthened the cytotoxicity of IR on SCC15-S cells in ferroptosis-dependent manner. The regulated GPX4/ferroptosis mediated the HBO function on re-sensitizing the radio-resistant OSCC cells to IR. In xenograft mice, co-exposure of IR and HBO can significantly reduce the tumor under IR activation compared with IR alone. Clinical data indicated that high GPX4 levels were associated with poor chemo-radiotherapy outcome. CONCLUSIONS HBO could re-sensitize radio-resistant OSCC cells through GPX4/ferroptosis regulation. These results provide a potential therapeutic strategy for clinical radio-resistance.
Collapse
Affiliation(s)
- Jia Liu
- Stomatology center, Shanxi Provincial People's hospital, Taiyuan, China
| | - Wei An
- Stomatology center, Shanxi Provincial People's hospital, Taiyuan, China
| | - Qian Zhao
- Stomatology center, Shanxi Provincial People's hospital, Taiyuan, China
| | - Zhen Liu
- The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Ying Jiang
- The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Huiqing Li
- Department of Anesthesiology, Shandong Provincial Third Hospital, Jinan, China
| | - Di Wang
- The Eighth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Avgoustakis K, Angelopoulou A. Biomaterial-Based Responsive Nanomedicines for Targeting Solid Tumor Microenvironments. Pharmaceutics 2024; 16:179. [PMID: 38399240 PMCID: PMC10892652 DOI: 10.3390/pharmaceutics16020179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Solid tumors are composed of a highly complex and heterogenic microenvironment, with increasing metabolic status. This environment plays a crucial role in the clinical therapeutic outcome of conventional treatments and innovative antitumor nanomedicines. Scientists have devoted great efforts to conquering the challenges of the tumor microenvironment (TME), in respect of effective drug accumulation and activity at the tumor site. The main focus is to overcome the obstacles of abnormal vasculature, dense stroma, extracellular matrix, hypoxia, and pH gradient acidosis. In this endeavor, nanomedicines that are targeting distinct features of TME have flourished; these aim to increase site specificity and achieve deep tumor penetration. Recently, research efforts have focused on the immune reprograming of TME in order to promote suppression of cancer stem cells and prevention of metastasis. Thereby, several nanomedicine therapeutics which have shown promise in preclinical studies have entered clinical trials or are already in clinical practice. Various novel strategies were employed in preclinical studies and clinical trials. Among them, nanomedicines based on biomaterials show great promise in improving the therapeutic efficacy, reducing side effects, and promoting synergistic activity for TME responsive targeting. In this review, we focused on the targeting mechanisms of nanomedicines in response to the microenvironment of solid tumors. We describe responsive nanomedicines which take advantage of biomaterials' properties to exploit the features of TME or overcome the obstacles posed by TME. The development of such systems has significantly advanced the application of biomaterials in combinational therapies and in immunotherapies for improved anticancer effectiveness.
Collapse
Affiliation(s)
- Konstantinos Avgoustakis
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece;
- Clinical Studies Unit, Biomedical Research Foundation Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece
| | - Athina Angelopoulou
- Department of Chemical Engineering, Polytechnic School, University of Patras, 26504 Patras, Greece
| |
Collapse
|
17
|
Fang R, Li Y, Jin J, Yang F, Chen J, Zhang J. Development of Anticancer Ferric Complex Based on Human Serum Albumin Nanoparticles That Generate Oxygen in Cells to Overcome Hypoxia-Induced Resistance in Metal Chemotherapy. J Med Chem 2024; 67:1184-1196. [PMID: 38181502 DOI: 10.1021/acs.jmedchem.3c01655] [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: 01/07/2024]
Abstract
To achieve the remarkable therapeutic efficacy of a ferric (Fe) complex via a reactive oxygen species (ROS) mechanism in solid tumors, a therapeutic Fe-based Schiff-base complex (Fe1) was synthesized and encapsulated in human serum albumin (HSA) nanoparticles (NPs), which generated oxygen (O2) in cancer cells in situ. The HSA-Fe1-O2 NP (HSA-Fe1-O2NP) delivery system effectively overcame hypoxia-induced resistance in metal chemotherapy, alleviated the hypoxic condition of tumor tissues, and showed excellent tumor suppression by generating excess ROS and promoting the apoptosis of SK-N-MC tumor cells. The HSA-Fe1-O2NPs not only enhanced the ability of the Fe1 complex to target tumor cells but also decreased adverse effects in vivo.
Collapse
Affiliation(s)
- Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North Second Road 109, Guilin, Guangxi 541004, P. R. China
| | - Yanping Li
- School of Public Health, Guilin Medical University, Huan Cheng North Second Road 109, Guilin, Guangxi 541004, P. R. China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North Second Road 109, Guilin, Guangxi 541004, P. R. China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North Second Road 109, Guilin, Guangxi 541004, P. R. China
| | - Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North Second Road 109, Guilin, Guangxi 541004, P. R. China
| |
Collapse
|
18
|
Bhatt K, Nukovic A, Colombani T, Bencherif SA. Biomaterial-assisted local oxygenation safeguards the prostimulatory phenotype and functions of human dendritic cells in hypoxia. Front Immunol 2023; 14:1278397. [PMID: 38169677 PMCID: PMC10758617 DOI: 10.3389/fimmu.2023.1278397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024] Open
Abstract
Dendritic cells (DCs), professional antigen-presenting cells, function as sentinels of the immune system. DCs initiate and fine-tune adaptive immune responses by presenting antigenic peptides to B and T lymphocytes to mount an effective immune response against cancer and pathogens. However, hypoxia, a condition characterized by low oxygen (O2) tension in different tissues, significantly impacts DC functions, including antigen uptake, activation and maturation, migration, as well as T-cell priming and proliferation. In this study, we employed O2-releasing biomaterials (O2-cryogels) to study the effect of localized O2 supply on human DC phenotype and functions. Our results indicate that O2-cryogels effectively mitigate DC exposure to hypoxia under hypoxic conditions. Additionally, O2-cryogels counteract hypoxia-induced inhibition of antigen uptake and migratory activity in DCs through O2 release and hyaluronic acid (HA) mediated mechanisms. Furthermore, O2-cryogels preserve and restore DC maturation and co-stimulation markers, including HLA-DR, CD86, and CD40, along with the secretion of proinflammatory cytokines in hypoxic conditions. Finally, our findings demonstrate that the supplemental O2 released from the cryogels preserves DC-mediated T-cell priming, ultimately leading to the activation and proliferation of allogeneic CD3+ T cells. This work emphasizes the potential of local oxygenation as a powerful immunomodulatory agent to improve DC activation and functions in hypoxia, offering new approaches for cancer and infectious disease treatments.
Collapse
Affiliation(s)
- Khushbu Bhatt
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Alexandra Nukovic
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| |
Collapse
|
19
|
Duan H, Wang F, Xu W, Sheng G, Sun Z, Chu H. Recent advances in the nanoarchitectonics of metal-organic frameworks for light-activated tumor therapy. Dalton Trans 2023; 52:16085-16102. [PMID: 37814810 DOI: 10.1039/d3dt02725b] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Metal-organic frameworks (MOFs) have received extensive attention in tumor therapy because of their advantages, including large specific surface area, regular pore size, adjustable shape, and facile functionalization. MOFs are porous materials formed by the coordination bonding of metal clusters and organic ligands. This review summarized the most recent advancements in tumor treatment based on nMOFs. First, we discuss the classification of MOFs, which primarily include the series of isoreticular MOF (IRMOF), zeolitic imidazolate framework (ZIF), coordination pillared-layer (CPL), Materials of Institute Lavoisier (MIL), porous coordination network (PCN), University of Oslo (UiO) and Biological metal-organic frameworks (BioMOFs). Then, we discuss the use of nMOFs in antitumor therapy, including drug delivery strategies, photodynamic therapy (PDT), photothermal therapy (PTT), and combination therapy. Finally, the obstacles and opportunities in nMOFs are discussed.
Collapse
Affiliation(s)
- Huijuan Duan
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Fang Wang
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Weizhe Xu
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Gang Sheng
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Zhaogang Sun
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Hongqian Chu
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| |
Collapse
|
20
|
Yuen CM, Tsai HP, Tseng TT, Tseng YL, Lieu AS, Kwan AL, Chang AYW. Hyperbaric Oxygen Therapy Adjuvant Chemotherapy and Radiotherapy through Inhibiting Stemness in Glioblastoma. Curr Issues Mol Biol 2023; 45:8309-8320. [PMID: 37886967 PMCID: PMC10605823 DOI: 10.3390/cimb45100524] [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: 08/13/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and deadliest primary brain tumor in adults. Despite the advances in GBM treatment, outcomes remain poor, with a 2-year survival rate of less than 5%. Hyperbaric oxygen (HBO) therapy is an intermittent, high-concentration, short-term oxygen therapy used to increase cellular oxygen content. In this study, we evaluated the effects of HBO therapy, alone or combined with other treatment modalities, on GBM in vitro and in vivo. In the in vitro analysis, we used a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to assess the effects of HBO therapy alone, a colony formation assay to analyze the effects of HBO therapy combined with radiotherapy and with temozolomide (TMZ), and a neurosphere assay to assess GBM stemness. In the in vivo analysis, we used immunohistochemical staining and in vivo bioluminescence imaging to assess GBM stemness and the therapeutic effect of HBO therapy alone or combined with TMZ or radiotherapy, respectively. HBO therapy did not affect GBM cell viability, but it did reduce the analyzed tumors' ability to form cancer stem cells. In addition, HBO therapy increased GBM sensitivity to TMZ and radiotherapy both in vitro and in vivo. HBO therapy did not enhance tumor growth and exhibited adjuvant effects to chemotherapy and radiotherapy through inhibiting GBM stemness. In conclusion, HBO therapy shows promise as an adjuvant treatment for GBM by reducing cancer stem cell formation and enhancing sensitivity to chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Chun-Man Yuen
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan;
- Division of Neurosurgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-P.T.); (T.-T.T.); (A.-S.L.)
| | - Tzu-Ting Tseng
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-P.T.); (T.-T.T.); (A.-S.L.)
| | - Yu-Lung Tseng
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 333, Taiwan;
| | - Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-P.T.); (T.-T.T.); (A.-S.L.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Aij-Lie Kwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-P.T.); (T.-T.T.); (A.-S.L.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22904, USA
| | - Alice Y. W. Chang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan;
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Cheng-Hsing Campus, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
21
|
Halma MTJ, Tuszynski JA, Marik PE. Cancer Metabolism as a Therapeutic Target and Review of Interventions. Nutrients 2023; 15:4245. [PMID: 37836529 PMCID: PMC10574675 DOI: 10.3390/nu15194245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Cancer is amenable to low-cost treatments, given that it has a significant metabolic component, which can be affected through diet and lifestyle change at minimal cost. The Warburg hypothesis states that cancer cells have an altered cell metabolism towards anaerobic glycolysis. Given this metabolic reprogramming in cancer cells, it is possible to target cancers metabolically by depriving them of glucose. In addition to dietary and lifestyle modifications which work on tumors metabolically, there are a panoply of nutritional supplements and repurposed drugs associated with cancer prevention and better treatment outcomes. These interventions and their evidentiary basis are covered in the latter half of this review to guide future cancer treatment.
Collapse
Affiliation(s)
- Matthew T. J. Halma
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- EbMC Squared CIC, Bath BA2 4BL, UK
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, I-1029 Turin, Italy
| | - Paul E. Marik
- Frontline COVID-19 Critical Care Alliance, Washington, DC 20036, USA
| |
Collapse
|
22
|
Hao J, Song Z, Su J, Li L, Zou L, Zou K. The PRX-1/TLR4 axis promotes hypoxia-induced radiotherapy resistance in non-small cell lung cancer by targeting the NF-κB/p65 pathway. Cell Signal 2023; 110:110806. [PMID: 37468052 DOI: 10.1016/j.cellsig.2023.110806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/29/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Hypoxic lung cancer cells are highly resistant to radiation. Peroxiredoxin-1 (PRX-1), a transcriptional coactivator that enhances the DNA-binding activity of serum reactive factor, has been identified as a target for radiotherapy sensitization, but the underlying molecular mechanism remains unclear. This study aimed to investigate the influence of PRX-1 on radiotherapy sensitivity in hypoxic tumors. Hypoxic lung cancer cells exhibited radiotherapy-resistant phenotypes after irradiation, including increased proliferation, DNA damage repair, cell migration, invasion and stemness. Radio-resistant hypoxic lung cancer cells showed high expression levels of PRX-1. Furthermore, we observed that PRX-1 bound to the promoter region of TRL4 (-300 to -600) and promoted its transcription and expression and that PRX-1/TRL4 activated the NF-κB/p65 signaling pathway. Increased radiotherapy resistance of hypoxic lung cancer cells increased their ability to proliferate, migrate, and maintain stemness in vivo and in vitro. These findings suggest that PRX-1/TRL4 could be used as a target for the treatment of radiotherapy-resistant lung cancer cells and further provide a theoretical basis for the clinical treatment of hypoxic lung cancer cells.
Collapse
Affiliation(s)
- Jiaojiao Hao
- The First Affiliated Hospital, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhuo Song
- The First Affiliated Hospital, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jiayi Su
- The First Affiliated Hospital, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Longjie Li
- The First Affiliated Hospital, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Lijian Zou
- The First Affiliated Hospital, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Kun Zou
- The First Affiliated Hospital, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
23
|
Guo S, Gu D, Yang Y, Tian J, Chen X. Near-infrared photodynamic and photothermal co-therapy based on organic small molecular dyes. J Nanobiotechnology 2023; 21:348. [PMID: 37759287 PMCID: PMC10523653 DOI: 10.1186/s12951-023-02111-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Near-infrared (NIR) organic small molecule dyes (OSMDs) are effective photothermal agents for photothermal therapy (PTT) due to their advantages of low cost and toxicity, good biodegradation, and strong NIR absorption over a wide wavelength range. Nevertheless, OSMDs have limited applicability in PTT due to their low photothermal conversion efficiency and inadequate destruction of tumor regions that are nonirradiated by NIR light. However, they can also act as photosensitizers (PSs) to produce reactive oxygen species (ROS), which can be further eradicated by using ROS-related therapies to address the above limitations of PTT. In this review, the synergistic mechanism, composition, and properties of photodynamic therapy (PDT)-PTT nanoplatforms were comprehensively discussed. In addition, some specific strategies for further improving the combined PTT and PDT based on OSMDs for cancer to completely eradicate cancer cells were outlined. These strategies include performing image-guided co-therapy, enhancing tumor infiltration, increasing H2O2 or O2 in the tumor microenvironment, and loading anticancer drugs onto nanoplatforms to enable combined therapy with phototherapy and chemotherapy. Meanwhile, the intriguing prospects and challenges of this treatment modality were also summarized with a focus on the future trends of its clinical application.
Collapse
Affiliation(s)
- Shuang Guo
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Dongyu Gu
- College of Marine Science and Environment, Dalian Ocean University, Dalian, 116023, China
| | - Yi Yang
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Jing Tian
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
24
|
Alpuim Costa D, Gonçalves-Nobre JG, Sampaio-Alves M, Guerra N, Arana Ribeiro J, Espiney Amaro C. Hyperbaric oxygen therapy as a complementary treatment in neuroblastoma - a narrative review. Front Oncol 2023; 13:1254322. [PMID: 37823059 PMCID: PMC10562625 DOI: 10.3389/fonc.2023.1254322] [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: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Neuroblastoma is the most frequently diagnosed cancer during the first year of life. This neoplasm originates from neural crest cells derived from the sympathetic nervous system, adrenal medulla, or paraspinal ganglia. The clinical presentation can vary from an asymptomatic mass to symptoms resulting from local invasion and/or spread of distant disease spread. The natural history of neuroblastoma is highly variable, ranging from relatively indolent biological behavior to a high-risk clinical phenotype with a dismal prognosis. Age, stage, and biological features are important prognostic risk stratification and treatment assignment prognostic factors. The multimodal therapy approach includes myeloablative chemotherapy, radiotherapy, immunotherapy, and aggressive surgical resection. Hyperbaric oxygen therapy (HBOT) has been proposed as a complementary measure to overcome tumor hypoxia, which is considered one of the hallmarks of this cancer treatment resistance. This article aims to review the relevant literature on the neuroblastoma pathophysiology, clinical presentation, and different biological and genetic profiles, and to discuss its management, focusing on HBOT.
Collapse
Affiliation(s)
- Diogo Alpuim Costa
- Hematology and Oncology Department, CUF Oncologia, Lisbon, Portugal
- Centro de Medicina Subaquática e Hiperbárica (CMSH), Portuguese Navy, Lisbon, Portugal
- Medical Oncology Department, Hospital de Cascais Dr. José de Almeida, Alcabideche, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade NOVA de Lisboa, Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - J. Guilherme Gonçalves-Nobre
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Hospital Garcia de Orta (HGO), E.P.E., Almada, Portugal
- Instituto de Saúde Ambiental (ISAMB), Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- Instituto de Medicina Preventiva & Saúde Pública (IMP&SP), Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- PTSurg – Portuguese Surgical Research Collaborative, Lisboa, Portugal PTSurg – Portuguese Surgical Research Collaborative, Lisbon, Portugal
| | - Mafalda Sampaio-Alves
- PTSurg – Portuguese Surgical Research Collaborative, Lisboa, Portugal PTSurg – Portuguese Surgical Research Collaborative, Lisbon, Portugal
- Faculty of Medicine, University of Porto, Oporto, Portugal
| | - Nuno Guerra
- Centro de Medicina Subaquática e Hiperbárica (CMSH), Portuguese Navy, Lisbon, Portugal
| | | | - Carla Espiney Amaro
- Centro de Medicina Subaquática e Hiperbárica (CMSH), Portuguese Navy, Lisbon, Portugal
| |
Collapse
|
25
|
Wang P, Wang XY, Man CF, Gong DD, Fan Y. Advances in hyperbaric oxygen to promote immunotherapy through modulation of the tumor microenvironment. Front Oncol 2023; 13:1200619. [PMID: 37790761 PMCID: PMC10543083 DOI: 10.3389/fonc.2023.1200619] [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: 04/05/2023] [Accepted: 08/25/2023] [Indexed: 10/05/2023] Open
Abstract
Hyperbaric oxygen therapy is a relatively safe treatment method that has been used for a long time in the clinic. It has been proven that it can enhance the sensitivity of radiotherapy and photodynamic therapy for cancer. However, there are few studies on hyperbaric oxygen and immunotherapy. In this article, we summarize that hyperbaric oxygen therapy regulates the tumor microenvironment through various pathways such as improving tumor hypoxia, targeting hypoxia-inducing factors, and generating reactive oxygen species. The change in the tumor microenvironment ultimately affects the curative effect of immunotherapy. Therefore, hyperbaric oxygen can influence immunotherapy by regulating the tumor microenvironment, providing a direction for the future development of immunotherapy.
Collapse
Affiliation(s)
- Pei Wang
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiao-Yan Wang
- Department of Gastroenterology, The Affiliated Suqian First People’s Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Chang-Feng Man
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dan-Dan Gong
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Fan
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
26
|
Nasr HY, Rifkin WJ, Muller JN, Chiu ES. Hyperbaric Oxygen Therapy for Threatened Nipple-Sparing Mastectomy Flaps: An Adjunct for Flap Salvage. Ann Plast Surg 2023; 90:S125-S129. [PMID: 36913565 DOI: 10.1097/sap.0000000000003441] [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: 03/14/2023]
Abstract
BACKGROUND Nipple-sparing mastectomy (NSM) is emerging as the standard of care for treatment of breast cancer because of its oncologic safety and superior aesthetic outcomes. However, ischemia or necrosis of the skin flap and/or nipple-areola complex remain frequent complications. Hyperbaric oxygen therapy (HBOT) has emerged as a potential adjunct for flap salvage, although it is not currently a widely accepted practice. Here we review our institution's experience using a protocol of HBOT in patients with signs of flap ischemia or necrosis after NSM. METHODS Retrospective review identified all patients treated with HBOT at our institution's hyperbaric and wound care center because of signs of ischemia after NSM. Treatment parameters consisted of 90-minute dives at 2.0 atmosphere once or twice daily. Patients unable to tolerate dives were considered a treatment failure, whereas those lost to follow-up were excluded from analysis. Patient demographics, surgical characteristics, and treatment indications were recorded. Primary outcomes assessed were flap salvage (no operative revision), need for revision procedures, and treatment complications. RESULTS A total of 17 patients and 25 breasts met the inclusion criteria. The mean ± SD time to initiation of HBOT was 9.47 ± 12.7 days. The mean ± SD age was 46.7 ± 10.4 years, and mean ± SD follow-up time was 36.5 ± 25.6 days. Indications for NSM included invasive cancer (41.2%), carcinoma in situ (29.4%), and breast cancer prophylaxis (29.4%). Initial reconstruction included tissue-expander placement (47.1%), autologous reconstruction with deep inferior epigastric flaps (29.4%), and direct-to-implant reconstruction (23.5%). Hyperbaric oxygen therapy indications included ischemia or venous congestion for 15 breasts (60.0%) and partial thickness necrosis for 10 breasts (40.0%). Flap salvage was achieved in 22 of 25 breasts (88.0%). Reoperation was required for 3 breasts (12.0%). Hyperbaric oxygen therapy-related complications were observed in 4 patients (23.5%), which included 3 patients with mild ear pain and 1 patient with severe sinus pressure leading to treatment abortion. CONCLUSIONS Nipple-sparing mastectomy is an invaluable tool for breast and plastic surgeons to achieve oncologic and cosmetic goals. However, ischemia or necrosis of the nipple-areola complex or mastectomy skin flap remains frequent complications. Hyperbaric oxygen therapy has emerged as a possible intervention for threatened flaps. Our results demonstrate the utility of HBOT in this population to achieve excellent NSM flap salvage rates.
Collapse
Affiliation(s)
- Hani Y Nasr
- From the Kimmel Hyperbaric and Advanced Wound Healing Center, Hansjörg Wyss Department of Plastic Surgery, NYU Langone Health, New York, NY
| | | | | | | |
Collapse
|
27
|
Zhou D, Fu D, Yan L, Xie L. The Role of Hyperbaric Oxygen Therapy in the Treatment of Surgical Site Infections: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:762. [PMID: 37109720 PMCID: PMC10145168 DOI: 10.3390/medicina59040762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/12/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
Surgical site infections (SSIs) are among the most prevalent postoperative complications, with significant morbidity and mortality worldwide. In the past half century, hyperbaric oxygen therapy (HBOT), the administration of 100% oxygen intermittently under a certain pressure, has been used as either a primary or alternative therapy for the management or treatment of chronic wounds and infections. This narrative review aims to gather information and evidence supporting the role of HBOT in the treatment of SSIs. We followed the Scale for the Quality Assessment of Narrative Review Articles (SANRA) guidelines and scrutinized the most relevant studies identified in Medline (via PubMed), Scopus, and Web of Science. Our review indicated that HBOT can result in rapid healing and epithelialization of various wounds and has potential beneficial effects in the treatment of SSIs or other similar infections following cardiac, neuromuscular scoliosis, coronary artery bypass, and urogenital surgeries. Moreover, it was a safe therapeutic procedure in most cases. The mechanisms related to the antimicrobial activity of HBOT include direct bactericidal effects through the formation of reactive oxygen species (ROS), the immunomodulatory effect of HBOT that increase the antimicrobial effects of the immune system, and the synergistic effects of HBOT with antibiotics. We emphasized the essential need for further studies, especially randomized clinical trials and longitudinal studies, to better standardize HBOT procedures as well as to determine its full benefits and possible side effects.
Collapse
Affiliation(s)
| | | | | | - Linshen Xie
- West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Zu Y, Wang Z, Yao H, Yan L. Oxygen-generating biocatalytic nanomaterials for tumor hypoxia relief in cancer radiotherapy. J Mater Chem B 2023; 11:3071-3088. [PMID: 36920849 DOI: 10.1039/d2tb02751h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Radiotherapy (RT), the most commonly used treatment method in clinics, shows unique advantages such as strong penetration, high energy intensity, and low systemic side effects. However, in vivo tumor hypoxia seriously hinders the therapeutic effect of RT. Hypoxia is a common characteristic of locally advanced solid tumor microenvironments, which leads to the proliferation, invasion and metastasis of tumor cells. In addition, oxygen consumption during RT will further aggravate tumor hypoxia, causing a variety of adverse side effects. In recent years, various biocatalytic nanomaterials (BCNs) have been explored to regulate and reverse tumor hypoxia microenvironments during RT. In this review, the most recent efforts toward developing oxygen-generating BCNs in relieving tumor hypoxia in RT are focused upon. The classification, engineering nanocatalytical activity of oxygen-generating BCNs and combined therapy based on these BCNs are systematically introduced and discussed. The challenges and prospects of these oxygen-generating BCNs in RT applications are also summarized.
Collapse
Affiliation(s)
- Yan Zu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.
| | - Ziyu Wang
- College of Medical and Biological lnformation Engineering, Northeastern University, Shenyang 110170, China
| | - Huiqin Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| | - Liang Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
29
|
Ingle J, Basu S. Mitochondria Targeted AIE Probes for Cancer Phototherapy. ACS OMEGA 2023; 8:8925-8935. [PMID: 36936289 PMCID: PMC10018722 DOI: 10.1021/acsomega.3c00203] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/20/2023] [Indexed: 06/01/2023]
Abstract
In recent years, mitochondrion (powerhouse of the cells) gained lots of interest as one of the unorthodox targets for futuristic cancer therapy. As a result, novel small molecules were developed to damage and image mitochondria in cancer models. In this context, aggregation-induced emission probes (AIEgens) received immense attention due to their applications in mitochondria-targeted biosensing, imaging, and biomedical theranostics. On the other hand, phototherapy (photodynamic and photothermal) has emerged as a powerful alternative to manage cancer due to its less invasive nature. However, merging these two areas to engineer mitochondria-targeted phototherapeutic probes for cancer diagnosis and treatment has remained a major challenge. In this mini-review, we will outline the development of novel mitochondria-targeted small molecule AIEgens as imaging agents and photosensitizers for photodynamic therapy along with dual photodymanic-phototheramal therapy and chemo-photodynamic therapy. We will also highlight the current challenges in developing mitochondria-targeted photothermal therapy probes for future biomedical theranostic applications to manage cancer.
Collapse
|
30
|
Teng KX, Niu LY, Yang QZ. Supramolecular Photosensitizer Enables Oxygen-Independent Generation of Hydroxyl Radicals for Photodynamic Therapy. J Am Chem Soc 2023; 145:4081-4087. [PMID: 36779824 DOI: 10.1021/jacs.2c11868] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
The highly oxygen-dependent nature of photodynamic therapy (PDT) limits its therapeutic efficacy against hypoxic solid tumors in clinics, which is an urgent problem to be solved. Herein, we develop an oxygen-independent supramolecular photodynamic agent that produces hydroxyl radical (•OH) by oxidizing water in the presence of intracellularly abundant pyruvic acid under oxygen-free conditions. A fluorene-substituted BODIPY was designed as the electron donor and coassembled with perylene diimide as the electron acceptor to form the quadruple hydrogen-bonded supramolecular photodynamic agent. Detailed mechanism studies reveal that intermolecular electron transfer and charge separation upon light irradiation result in an efficient generation of radical ion pairs. Under oxygen-free conditions, the cationic radicals directly oxidize water to generate highly cytotoxic •OH, and the anionic radicals transfer electrons to pyruvic acid, realizing the catalytic cycle. Thus, this photodynamic agent exhibited superb photocytotoxicity even under severe hypoxic environments and excellent in vivo antitumor efficacy on HeLa-bearing mouse models. This work provides a strategy for constructing oxygen-independent photodynamic agents, which opens up an avenue for effective PDT against hypoxic tumors.
Collapse
Affiliation(s)
- Kun-Xu Teng
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Li-Ya Niu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing-Zheng Yang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
31
|
Anti-Hypoxia Nanoplatforms for Enhanced Photosensitizer Uptake and Photodynamic Therapy Effects in Cancer Cells. Int J Mol Sci 2023; 24:ijms24032656. [PMID: 36768975 PMCID: PMC9916860 DOI: 10.3390/ijms24032656] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Photodynamic therapy (PDT) holds great promise in cancer eradication due to its target selectivity, non-invasiveness, and low systemic toxicity. However, due to the hypoxic nature of many native tumors, PDT is frequently limited in its therapeutic effect. Additionally, oxygen consumption during PDT may exacerbate the tumor's hypoxic condition, which stimulates tumor proliferation, metastasis, and invasion, resulting in poor treatment outcomes. Therefore, various strategies have been developed to combat hypoxia in PDT, such as oxygen carriers, reactive oxygen supplements, and the modulation of tumor microenvironments. However, most PDT-related studies are still conducted on two-dimensional (2D) cell cultures, which fail to accurately reflect tissue complexity. Thus, three-dimensional (3D) cell cultures are ideal models for drug screening, disease simulation and targeted cancer therapy, since they accurately replicate the tumor tissue architecture and microenvironment. This review summarizes recent advances in the development of strategies to overcome tumor hypoxia for enhanced PDT efficiency, with a particular focus on nanoparticle-based photosensitizer (PS) delivery systems, as well as the advantages of 3D cell cultures.
Collapse
|
32
|
Claiborne MD. Manipulation of metabolic pathways to promote stem-like and memory T cell phenotypes for immunotherapy. Front Immunol 2023; 13:1061411. [PMID: 36741362 PMCID: PMC9889361 DOI: 10.3389/fimmu.2022.1061411] [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: 10/04/2022] [Accepted: 12/21/2022] [Indexed: 01/20/2023] Open
Abstract
Utilizing the immune system's capacity to recognize and kill tumor cells has revolutionized cancer therapy in recent decades. Phenotypic study of antitumor T cells supports the principle that superior tumor control is achieved by cells with more long-lived memory or stem-like properties as compared to terminally differentiated effector cells. In this Mini-Review, we explore recent advances in profiling the different metabolic programs that both generate and define subsets of memory T cells. We additionally discuss new experimental approaches that aim to maximize the durability and sustained antitumor response associated with memory T cells within the unique immunosuppressive conditions of the tumor microenvironment, such as engineered attempts to overcome hypoxia-induced changes in mitochondrial function, the inhibitory effects of tumor metabolites, and exploitation of more recently-defined metabolic pathways controlling T cell memory fate such as glycogen metabolism.
Collapse
|
33
|
Wang C, Cheng T, Lu Q, Li W, Liu B, Yue L, Du M, Sheng W, Lu Z, Yang J, Geng F, Gao X, Lü J, Pan X. Oxygen therapy accelerates apoptosis induced by selenium compounds via regulating Nrf2/MAPK signaling pathway in hepatocellular carcinoma. Pharmacol Res 2023; 187:106624. [PMID: 36563868 DOI: 10.1016/j.phrs.2022.106624] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 12/25/2022]
Abstract
Selenium has good antitumor effects in vitro, but the hypoxic microenvironment in solid tumors makes its clinical efficacy unsatisfactory. We hypothesized that the combination with oxygen therapy might improve the treatment efficacy of selenium in hypoxic tumors through the changes of redox environment. In this work, two selenium compounds, Na2SeO3 and CysSeSeCys, were selected to interrogate their therapeutic effects on hepatocellular carcinoma (HCC) under different oxygen levels. In tumor-bearing mice, both selenium compounds significantly inhibited the tumor growth, and combined with oxygen therapy further reduced the tumor volume about 50 %. In vitro HepG2 cell experiments, selenium induced autophagy and delayed apoptosis under hypoxia (1 % O2), while inhibited autophagy and accelerated apoptosis under hyperoxia (60 % O2). We found that, in contrast to hypoxia, the hyperoxic environment facilitated the H2Se, produced by the selenium metabolism in cells, to be rapidly oxidized to generate H2O2, leading to inhibit the expression level of Nrf2 and to increase that of phosphorylation of p38 and MKK4, resulting in inhibiting autophagy and accelerating apoptosis. Once the Nrf2 gene was knocked down, selenium compounds combined with hyperoxia treatment would further activate the MAPK signaling pathway and further increase apoptosis. These findings highlight oxygen can significantly enhance the anti-HCC effect of selenium compounds through regulating the Nrf2 and MAPK signaling pathways, thus providing novel therapeutic strategy for the hypoxic tumors and pave the way for the application of selenium in clinical treatment.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | | | - Qianqian Lu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264003, China
| | - Wenzhen Li
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Ben Liu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264003, China
| | - Lijun Yue
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Maoru Du
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Wenxue Sheng
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Zhaochen Lu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Jingnan Yang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Feng Geng
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Xue Gao
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| | - Junhong Lü
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250000, China.; Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Xiaohong Pan
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
34
|
Adebayo AK, Nakshatri H. Modeling Preclinical Cancer Studies under Physioxia to Enhance Clinical Translation. Cancer Res 2022; 82:4313-4321. [PMID: 36169928 PMCID: PMC9722631 DOI: 10.1158/0008-5472.can-22-2311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/31/2022] [Accepted: 09/23/2022] [Indexed: 01/24/2023]
Abstract
Oxygen (O2) plays a key role in cellular homeostasis. O2 levels are tightly regulated in vivo such that each tissue receives an optimal amount to maintain physiologic status. Physiologic O2 levels in various organs range between 2% and 9% in vivo, with the highest levels of 9% in the kidneys and the lowest of 0.5% in parts of the brain. This physiologic range of O2 tensions is disrupted in pathologic conditions such as cancer, where it can reach as low as 0.5%. Regardless of the state, O2 tension in vivo is maintained at significantly lower levels than ambient O2, which is approximately 21%. Yet, routine in vitro cellular manipulations are carried out in ambient air, regardless of whether or not they are eventually transferred to hypoxic conditions for subsequent studies. Even brief exposure of hematopoietic stem cells to ambient air can cause detrimental effects through a mechanism termed extraphysiologic oxygen shock/stress (EPHOSS), leading to reduced engraftment capabilities. Here, we provide an overview of the effects of ambient air exposure on stem and non-stem cell subtypes, with a focus on recent findings that reveal the impact of EPHOSS on cancer cells.
Collapse
Affiliation(s)
- Adedeji K. Adebayo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
35
|
Klitgaard TL, Schjørring OL, Severinsen MT, Perner A, Rasmussen BS. Lower versus higher oxygenation targets in ICU patients with haematological malignancy - insights from the HOT-ICU trial. BJA OPEN 2022; 4:100090. [PMID: 37588787 PMCID: PMC10430820 DOI: 10.1016/j.bjao.2022.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/14/2022] [Indexed: 08/18/2023]
Abstract
Background Patients admitted to an intensive care unit (ICU) with active haematological malignancy and hypoxaemic respiratory failure have a high mortality. Oxygen supplementation is essential, but limited information exists on the optimum oxygenation targets in these patients. Methods This subgroup analysis was specified before completion of the Handling Oxygenation Targets in the ICU (HOT-ICU) trial. The trial investigated the effects of a lower (8 kPa) vs a higher (12 kPa) arterial oxygenation target and was stratified for active haematological malignancy, chronic obstructive pulmonary disease, and site. We here report the primary outcome (90-day mortality) and selected secondary outcomes in the subgroup of patients with active haematological malignancy. Results The HOT-ICU trial included 168 patients with active haematological malignancy; 82 were randomly allocated to an arterial oxygenation target of 8 kPa, and 86 to 12 kPa. At 90 days, 53/81 patients (65%) in the lower-oxygenation group and 47/86 patients (55%) in the higher-oxygenation group had died: adjusted relative risk 1.22 (95% confidence interval 0.95-1.56); at 1 year, the numbers were 58/81 (72%) vs 56/86 (65%): adjusted relative risk 1.11 (95% confidence interval 0.90-1.36). No statistically significant differences were found for any secondary outcomes. Conclusion In ICU patients with active haematological malignancies and hypoxaemic respiratory failure, we found a high mortality at 90 days and 1 year. Our results did not preclude clinically relevant benefits or harms of a lower oxygenation target in patients with active haematological malignancy. A randomised trial may, therefore, be worthwhile for these patients. Clinical trial registration NCT03174002.
Collapse
Affiliation(s)
- Thomas L. Klitgaard
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Olav L. Schjørring
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Marianne T. Severinsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Research Centre, Aalborg University Hospital, Aalborg, Denmark
| | - Anders Perner
- Department of Intensive Care, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Bodil S. Rasmussen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
36
|
Salim SA, Salaheldin TA, Elmazar MM, Abdel-Aziz AF, Kamoun EA. Smart biomaterials for enhancing cancer therapy by overcoming tumor hypoxia: a review. RSC Adv 2022; 12:33835-33851. [PMID: 36505711 PMCID: PMC9693911 DOI: 10.1039/d2ra06036a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Hypoxia is a distinctive feature of most solid tumors due to insufficient oxygen supply of the abnormal vasculature, which cannot work with the demands of the fast proliferation of cancer cells. One of the main obstacles to limiting the efficacy of cancer medicines is tumor hypoxia. Thus, oxygen is a vital parameter for controlling the efficacy of different types of cancer therapy, such as chemotherapy (CT), photodynamic therapy (PDT), photothermal therapy (PTT), immunotherapy (IT), and radiotherapy (RT). Numerous technologies have attracted much attention for enhancing oxygen distribution in humans and improving the efficacy of cancer treatment. Such technologies include treatment with hyperbaric oxygen therapy (HBO), delivering oxygen by polysaccharides (e.g., cellulose, gelatin, alginate, and silk) and other biocompatible synthetic polymers (e.g., PMMA, PLA, PVA, PVP and PCL), decreasing oxygen consumption, producing oxygen in situ in tumors, and using polymeric systems as oxygen carriers. Herein, this review provides an overview of the relationship between hypoxia in tumor cells and its role in the limitation of different cancer therapies alongside the numerous strategies for oxygen delivery using polysaccharides and other biomaterials as carriers and for oxygen generation.
Collapse
Affiliation(s)
- Samar A. Salim
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE)El-Sherouk CityCairo 11837Egypt+20-1283320302,Biochemistry Group, Dep. of Chemistry, Faculty of Science, Mansoura UniversityEgypt
| | - Taher A. Salaheldin
- Department of Medicine, Case Western Reserve University School of MedicineClevelandOH44106USA
| | - Mohamed M. Elmazar
- Faculty of Pharmacy, The British University in Egypt (BUE)El-Sherouk CityCairo 11837Egypt
| | - A. F. Abdel-Aziz
- Biochemistry Group, Dep. of Chemistry, Faculty of Science, Mansoura UniversityEgypt
| | - Elbadawy A. Kamoun
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE)El-Sherouk CityCairo 11837Egypt+20-1283320302,Polymeric Materials Research Dep., Advanced Technology and New Materials Research Institute (ATNMRI), The City of Scientific Research and Technological Applications (SRTA-City)New Borg Al-Arab City 21934AlexandriaEgypt
| |
Collapse
|
37
|
Zhan Q, Han X, Mu J, Shi X, Zheng Y, Wang T, Cao T, Xi Y, Weng Z, Wang X, Cao P. Oxygen-evolving hollow polydopamine alleviates tumour hypoxia for enhancing photodynamic therapy in cancer treatment. NANOSCALE ADVANCES 2022; 4:5021-5026. [PMID: 36504744 PMCID: PMC9680955 DOI: 10.1039/d2na00549b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/03/2022] [Indexed: 06/17/2023]
Abstract
Hypoxia, a characteristic hallmark of solid tumours, restricts the therapeutic effect of photodynamic therapy (PDT) for cancer treatment. To address this issue, a facile and nanosized oxygen (O2) bubble template is established by mixing oxygenated water and water-soluble solvents for guiding hollow polydopamine (HPDA) synthesis, and O2 is encapsulated in the cavity of HPDA. HPDA with abundant catechol is designed as a carrier for zinc phthalocyanine (ZnPc, a boronic acid modified photosensitizer) via borate ester bonds to fabricate nanomedicine (denoted as HZNPs). The in vitro and in vivo results indicate that O2-evolving HZNPs could alleviate tumour hypoxia and enhance PDT-anticancer efficiency. Melanin-like HPDA with a photothermal conversion rate (η) of 38.2% shows excellent synergistic photothermal therapy (PTT) efficiency in cancer treatment.
Collapse
Affiliation(s)
- Qichen Zhan
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
| | - Xuan Han
- School of Chinese Medicine, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
| | - Jiankang Mu
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
| | - Xianqing Shi
- Department of Public Experimental Teaching, Nanjing University of Aeronautics and Astronautics Nanjing Jiangsu 211106 China
| | - Yuhan Zheng
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
| | - Ting Wang
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry Nanjing Jiangsu 210042 China
| | - Tao Cao
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
| | - Yulu Xi
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
| | - Zhongpei Weng
- Gaoyou Hospital of Traditional Chinese Medicine Yangzhou Jiangsu 225600 China
| | - Xiaoqing Wang
- Gaoyou Hospital of Traditional Chinese Medicine Yangzhou Jiangsu 225600 China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine Nanjing Jiangsu 210023 China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine Nanjing Jiangsu 210028 China
- Zhenjiang Hospital of Chinese Traditional and Western Medicine Zhenjiang Jiangsu 212002 China
| |
Collapse
|
38
|
Wu Y, Ding L, Zheng C, Li H, Wu M, Sun Y, Liu X, Zhang X, Zeng Y. Targeted co-delivery of a photosensitizer and an antisense oligonucleotide based on an activatable hyaluronic acid nanosystem with endogenous oxygen generation for enhanced photodynamic therapy of hypoxic tumors. Acta Biomater 2022; 153:419-430. [PMID: 36115655 DOI: 10.1016/j.actbio.2022.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022]
Abstract
Photodynamic therapy (PDT) is a promising cancer treatment modality with advantages of minimal invasiveness, repeatable therapy, and mild systemic toxicity. However, the limited bioavailability of photosensitizer (PS), tumor hypoxia, and the presence of antiapoptotic proteins in cancer cells, has hampered the efficiency of PDT. To address these limitations, herein, we developed a hyaluronic acid (HA) based nanosystem (HA-Ce6-Hemin@DNA-Protamine NPs, HCH@DP) loaded with chlorin e6 (Ce6, as PS), hemin (as mimetic catalase) and antisense oligonucleotide (ASO) of B-cell lymphoma 2 (Bcl-2) anti-apoptosis protein via a simple electrostatic self-assembly method for enhanced PDT of hypoxic solid tumors. The HCH@DP can target deliver the PS and ASO to tumor cells via cancer cell overexpressed HA receptors (i.e., CD44 or RHAMM). The Ce6 was released from HA-ss-Ce6 (HSC conjugates) after the reaction of cleavable disulfide bond with glutathione (GSH), which recovered the fluorescence and phototoxicity of Ce6 upon laser irradiation. Meanwhile, the catalase-mimicking hemin (degradation of HA-eda-hemin by hyaluronidase) decomposed the tumor overdressed endogenous H2O2 to oxygen, which relieved tumor hypoxia and further overcome hypoxia-associated resistance of PDT. Furthermore, the inhibition of Bcl-2 expression by Bcl-2 ASO also greatly improved the cellular sensitivity to PDT. Both in vitro and in vivo results showed the tumor cell targeting ability, hypoxia relief and significantly enhanced antitumor PDT efficacy of HCH@DP for hypoxic tumor cells upon laser irradiation. Thus, by improving the target delivery of PS and ASO, relieving tumor hypoxia, and down-regulation of anti-apoptotic proteins, this HCH@DP nanosystem achieved enhanced PDT efficiency against hypoxic tumors. In general, our work provided a promising strategy to increase the utilization of key components (PS and oxygen) of PDT and the cell sensitivity to PDT by targeting co-delivery PS and oligonucleotides to tumor cells via a biocompatible HA based carrier, thereby achieving efficiently PDT treatment of hypoxic solid tumors with potential translation possibility. STATEMENT OF SIGNIFICANCE: The efficiency of PDT against solid tumor is severely restricted by the limited bioavailability of photosensitizer, tumor hypoxia, and the presence of antiapoptotic proteins in cancer cells. Herein, we have developed an activatable hyaluronic acid (HA) based nanosystem (HA-Ce6-Hemin@DNA-Protamine NPs, HCH@DP) via a simple electrostatic self-assembly method for PDT treatment of hypoxic solid tumors. The HCH@DP enabled to target co-delivery of photosensitizer and antisense oligonucleotide to tumor cells, overcoming tumor hypoxia through in situ oxygen production and improving cellular sensitivity by efficiently reducing anti-apoptosis effect of cancer cells for synergistically enhancing PDT efficiency. This work suggests a promising strategy to develop small molecule drug and oligonucleotides co-delivery nanoplatforms for efficiently PDT treatment of hypoxic solid tumor.
Collapse
Affiliation(s)
- Yanni Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China
| | - Lei Ding
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341119, PR China
| | - Cheng Zheng
- College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Hongsheng Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China
| | - Yupeng Sun
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China
| | - Xiaolong Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China.
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, PR China; Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, PR China.
| |
Collapse
|
39
|
Tao Y, Liu Y, Dong Z, Chen X, Wang Y, Li T, Li J, Zang S, He X, Chen D, Zhao Z, Li M. Cellular Hypoxia Mitigation by Dandelion-like Nanoparticles for Synergistic Photodynamic Therapy of Oral Squamous Cell Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44039-44053. [PMID: 36153957 DOI: 10.1021/acsami.2c10021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Hypoxia at the tumor site limits the therapeutic effects of photodynamic therapy (PDT) in oral squamous cell carcinoma (OSCC), which is an oxygen-consumption process. Inhibiting cellular oxygen consumption and reducing cellular ATP production are expected to enhance PDT. In this study, we designed and constructed dandelion-like size-shrinkable nanoparticles for tumor-targeted delivery of hypoxia regulator resveratrol (RES) and photodynamic agent chlorine e6 (CE6). Both drugs were co-encapsulated in small-sized micelles modified with EGFR targeting ligand GE11, which was further conjugated on hyaluronic nanogel (NG) to afford RC-GMN. After targeted accumulation in tumors mediated by GE11 and enhanced penetration and retention (EPR) effects, RC-GMN was degraded by hyaluronidase (HAase) and resulted in small-sized micelles, allowing for deep penetration and dual-receptor-mediated cellular internalization. Resveratrol inhibited cellular oxygen consumption and provided sufficient oxygen for PDT, which consequently activated PDT to produce reactive oxygen species (ROS). Notably, we found that autophagy was overactivated in PDT, which was further strengthened by the hypoxia regulator resveratrol, elevating autophagic cell death. The synergistic effects of resveratrol and CE6 promoted autophagic cell death and apoptosis in the enhanced PDT, resulting in stronger antitumor effects in the orthotopic OSCC model. Therefore, the facilitated delivery of hypoxia regulator enhanced PDT efficacy by elevating oxygen content in tumor cells and inducing autophagic cell death and apoptosis, which offers an alternative strategy for enhancing the PDT effects against OSCC.
Collapse
Affiliation(s)
- Yuan Tao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yingke Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaoxiao Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yashi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ting Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Shuya Zang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xuan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Dong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
40
|
Chen G, Wu K, Li H, Xia D, He T. Role of hypoxia in the tumor microenvironment and targeted therapy. Front Oncol 2022; 12:961637. [PMID: 36212414 PMCID: PMC9545774 DOI: 10.3389/fonc.2022.961637] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
Tumor microenvironment (TME), which is characterized by hypoxia, widely exists in solid tumors. As a current research hotspot in the TME, hypoxia is expected to become a key element to break through the bottleneck of tumor treatment. More and more research results show that a variety of biological behaviors of tumor cells are affected by many factors in TME which are closely related to hypoxia. In order to inhibiting the immune response in TME, hypoxia plays an important role in tumor cell metabolism and anti-apoptosis. Therefore, exploring the molecular mechanism of hypoxia mediated malignant tumor behavior and therapeutic targets is expected to provide new ideas for anti-tumor therapy. In this review, we discussed the effects of hypoxia on tumor behavior and its interaction with TME from the perspectives of immune cells, cell metabolism, oxidative stress and hypoxia inducible factor (HIF), and listed the therapeutic targets or signal pathways found so far. Finally, we summarize the current therapies targeting hypoxia, such as glycolysis inhibitors, anti-angiogenesis drugs, HIF inhibitors, hypoxia-activated prodrugs, and hyperbaric medicine.
Collapse
Affiliation(s)
- Gaoqi Chen
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Kaiwen Wu
- Department of Gastroenterology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hao Li
- Deparment of Neurology, Affiliated Hospital of Jiangsu University, Jiang Su University, Zhenjiang, China
| | - Demeng Xia
- Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
- *Correspondence: Demeng Xia, ; Tianlin He,
| | - Tianlin He
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Demeng Xia, ; Tianlin He,
| |
Collapse
|
41
|
Xu XX, Chen SY, Yi NB, Li X, Chen SL, Lei Z, Cheng DB, Sun T. Research progress on tumor hypoxia-associative nanomedicine. J Control Release 2022; 350:829-840. [PMID: 36100192 DOI: 10.1016/j.jconrel.2022.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Hypoxia at the solid tumor site is generally related to the unrestricted proliferation and metabolism of cancerous cells, which can cause tumor metastasis and aggravate tumor progression. Besides, hypoxia plays a substantial role in tumor treatment, and it is one of the main reasons that malignant tumors are difficult to cure and have a poor prognosis. On account of the tumor specific hypoxic environment, many hypoxia-associative nanomedicine have been proposed for tumor treatment. Considering the enhanced targeting effect, designing hypoxia-associative nanomedicine can not only minimize the adverse effects of drugs on normal tissues, but also achieve targeted therapy at the lesion site. Mostly, there can be three strategies for the treatment of hypoxic tumor, including improvement of hypoxic environment, hypoxia responsive drug release and hypoxia activated prodrug. The review describes the design principle and applications of tumor hypoxia-associative nanomedicine in recent years, and also explores its development trends in solid tumor treatment. Moreover, this review presents the current limitations of tumor hypoxia-associative nanomedicine in chemotherapy, radiotherapy, photodynamic therapy, sonodynamic therapy and immunotherapy, which may provide a reference for clinic translation of tumor hypoxia-associative nanomedicine.
Collapse
Affiliation(s)
- Xiao-Xue Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Yi Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Ning-Bo Yi
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xin Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| |
Collapse
|
42
|
Sethuraman KN, Smolin R, Henry S. Is There a Place for Hyperbaric Oxygen Therapy? Adv Surg 2022; 56:169-204. [PMID: 36096567 DOI: 10.1016/j.yasu.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Hyperbaric oxygen therapy (HBOT) involves treating patients by providing 100% oxygen through inhalation while inside a treatment pressurized chamber. The oxygen acts as a drug and the hyperbaric chamber as the dosing device. The effect of hyperbaric hyperoxia is dose dependent and, therefore, treatment depth and duration are important when considering its use. HBOT can either be the primary method of treatment or used adjunctively to medications or surgical techniques. The underpinning physiology is to bring oxygen-rich plasma to hypoxic tissue, preventing reperfusion injury, strengthening immune responsiveness, and encouraging new collagen deposition as well as endothelial cell formation.
Collapse
Affiliation(s)
- Kinjal N Sethuraman
- University of Maryland Medical Center, Hyperbaric and Dive Medicine, 22 South Greene Street, Baltimore, MD 2120, USA
| | - Ryan Smolin
- University of Maryland School of Medicine, 685 West Baltimore Street, Suite 150, Baltimore, MD 21201, USA
| | - Sharon Henry
- University of Maryland Medical Center, R A Cowley Shock Trauma Center, Room T1R59, 22 South Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
43
|
Engineering micro oxygen factories to slow tumour progression via hyperoxic microenvironments. Nat Commun 2022; 13:4495. [PMID: 35918337 PMCID: PMC9345862 DOI: 10.1038/s41467-022-32066-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/18/2022] [Indexed: 12/11/2022] Open
Abstract
While hypoxia promotes carcinogenesis, tumour aggressiveness, metastasis, and resistance to oncological treatments, the impacts of hyperoxia on tumours are rarely explored because providing a long-lasting oxygen supply in vivo is a major challenge. Herein, we construct micro oxygen factories, namely, photosynthesis microcapsules (PMCs), by encapsulation of acquired cyanobacteria and upconversion nanoparticles in alginate microcapsules. This system enables a long-lasting oxygen supply through the conversion of external radiation into red-wavelength emissions for photosynthesis in cyanobacteria. PMC treatment suppresses the NF-kB pathway, HIF-1α production and cancer cell proliferation. Hyperoxic microenvironment created by an in vivo PMC implant inhibits hepatocarcinoma growth and metastasis and has synergistic effects together with anti-PD-1 in breast cancer. The engineering oxygen factories offer potential for tumour biology studies in hyperoxic microenvironments and inspire the exploration of oncological treatments.
Collapse
|
44
|
Pan H, Zou Q, Wang T, Li D, Sun SK. Minimalist O 2 generator formed by in situ KMnO 4 oxidation for tumor cascade therapy. Biomaterials 2022; 287:121596. [PMID: 35700623 DOI: 10.1016/j.biomaterials.2022.121596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/21/2022]
Abstract
Diverse oxygen generation strategies have been developed to overcome hypoxia in tumors for enhancing the therapeutic efficacy, but inevitably suffering from tedious synthesis process of oxygen generators in vitro before in vivo administration. Herein, we show direct injection of commercially and clinically used KMnO4 into solid tumors enables in situ formation of MnO2 as an oxygen depot for cascade oxidation damage and enhanced photodynamic therapy. KMnO4 can damage tumor tissues by oxidation and generate MnO2, and subsequent intravenous injection of Ce6 allows MnO2-triggered hypoxia-modulated photodynamic therapy of tumors. Excellent cascade tumor suppression effect is realized both in vitro and in vivo based on the KMnO4-Ce6 system without the need of synthesis. The proposed strategy lays down a novel way with unprecedented superiors of no need of synthesis process and ultra-facile administration procedure for tumor hypoxia-modulated cascade therapy.
Collapse
Affiliation(s)
- Haiyan Pan
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Tingting Wang
- Department of Ultrasound in Medicine, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, 310009, China
| | - Dong Li
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China.
| |
Collapse
|
45
|
Li K, Gong Y, Qiu D, Tang H, Zhang J, Yuan Z, Huang Y, Qin Y, Ye L, Yang Y. Hyperbaric oxygen facilitates teniposide-induced cGAS-STING activation to enhance the antitumor efficacy of PD-1 antibody in HCC. J Immunother Cancer 2022; 10:jitc-2021-004006. [PMID: 36002188 PMCID: PMC9413187 DOI: 10.1136/jitc-2021-004006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Emerging evidence indicates that the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) axis plays a pivotal role in intrinsic antitumor immunity. Previous studies demonstrate that the conventional chemotherapy agent, teniposide, effectively promotes the therapeutic efficacy of programmed cell death protein-1 antibody (PD-1 Ab) through robust cGAS-STING activation. Unfortunately, the cGAS expression of tumor cells is reported to be severely suppressed by the hypoxic status in solid tumor. Clinically, enhancing chemotherapy-induced, DNA-activated tumor STING signaling by alleviating tumor hypoxia might be one possible direction for improving the currently poor response rates of patients with hepatocellular carcinoma (HCC) to PD-1 Ab. METHODS Teniposide was first screened out from several chemotherapy drugs according to their potency in inducing cGAS-STING signaling in human HCC cells. Teniposide-treated HCC cells were then cultured under hypoxia, normoxia or reoxygenation condition to detect change in cGAS-STING signaling. Next, oxaliplatin/teniposide chemotherapy alone or combined with hyperbaric oxygen (HBO) therapy was administered on liver orthotopic mouse tumor models, after which the tumor microenvironment (TME) was surveyed. Lastly, teniposide alone or combined with HBO was performed on multiple mouse tumor models and the subsequent anti-PD-1 therapeutic responses were observed. RESULTS Compared with the first-line oxaliplatin chemotherapy, teniposide chemotherapy induced stronger cGAS-STING signaling in human HCC cells. Teniposide-induced cGAS-STING activation was significantly inhibited by hypoxia inducible factor 1α in an oxygen-deficient environment in vitro and the inhibition was rapidly removed via effective reoxygenation. HBO remarkably enhanced the cGAS-STING-dependent tumor type Ⅰ interferon and nuclear factor kappa-B signaling induced by teniposide in vivo, both of which contributed to the activation of dendritic cells and subsequent cytotoxic T cells. Combined HBO with teniposide chemotherapy improved the therapeutic effect of PD-1 Ab in multiple tumor models. CONCLUSIONS By combination of two therapies approved by the Food and Drug Administration, we safely stimulated an immunogenic, T cell-inflamed HCC TME, leading to further sensitization of tumors to anti-PD-1 immunotherapy. These findings might enrich therapeutic strategies for advanced HCC andwe can attempt to improve the response rates of patients with HCC to PD-1 Ab by enhancing DNA-activated STING signaling through effective tumor reoxygenation.
Collapse
Affiliation(s)
- Kun Li
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yihang Gong
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongbo Qiu
- Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Tang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zenan Yuan
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingqi Huang
- Department of Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yunfei Qin
- Department of Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linsen Ye
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Sun J, Wang J, Hu W, Wang Y, Zhang Q, Hu X, Chou T, Zhang B, Gallaro C, Halloran M, Liang L, Ren L, Wang H. A Porous Bimetallic Au@Pt Core-Shell Oxygen Generator to Enhance Hypoxia-Dampened Tumor Chemotherapy Synergized with NIR-II Photothermal Therapy. ACS NANO 2022; 16:10711-10728. [PMID: 35838683 DOI: 10.1021/acsnano.2c02528] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The characteristic hypoxia of solid tumors and inadequate oxygen supply become a key causation of the resistance to chemotherapy in cancer treatment. Herein, a bimetallic oxygen nanogenerator, i.e., porous Au@Pt core-shell nanostructures, is particularly developed to reduce the multidrug resistance by oxygenating the tumor along with synergistic chemo-photothermal therapy for efficient tumor eradication. The porous platinum (Pt) shell was able to catalyze oxygen generation from endogenous hydrogen peroxide in the tumor, reducing the exocytosis of doxorubicin (DOX) via suppressed expression of hypoxia-inducible factor-1α, multidrug resistance gene 1, and P-glycoprotein. The strong absorbance of Au@Pt nanostructures in NIR window II enabled NIR-II photothermal therapy. Further incorporation of DOX into the mesopores of Au@Pt nanostructures with the assistance of phase change materials (PCM) led to the formulation of Au@Pt-DOX-PCM-PEG nanotherapeutics for NIR-II-activated chemotherapy. This work presents an efficient H2O2-driven oxygenerator for enhanced hypoxia-dampened chemotherapy and NIR-II photothermal therapy.
Collapse
Affiliation(s)
- Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, People's Republic of China
| | - Wei Hu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Yuhao Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Qiang Zhang
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian 361005, People's Republic of China
| | - Xiaotong Hu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Tsengming Chou
- Laboratory for Multiscale Imaging, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Cosmo Gallaro
- Department of Physics, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, New Jersey 07030, United States
| | - Meghan Halloran
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Lyu Liang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian 361005, People's Republic of China
| | - Hongjun Wang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
- Center for Healthcare Innovation, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, New Jersey 07030, United States
| |
Collapse
|
47
|
Zhang H, Yan X, Zhang Y, Bao C, Li C. An oxygen-economical nano-photosensitizer with a high photodynamic therapeutic outcome via simultaneous reduction of the cellular respiration and oxygen depletion of PDT. J Mater Chem B 2022; 10:4623-4631. [PMID: 35647782 DOI: 10.1039/d2tb00309k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of photodynamic nanomedicines that can alleviate intratumoral oxygen deficiency during photodynamic therapy (PDT) is of great significance for improving the therapeutic outcome of solid tumors characterized by severe hypoxia. Massive oxygen consumption due to vigorous cellular respiration, i.e., mitochondrial-associated oxidative phosphorylation (OXPHOS), is another major cause of severe tumor hypoxia in addition to insufficient oxygen supply. Moreover, oxygen depletion during PDT further exacerbates the shortage of intratumoral oxygen. In this work, we engineered a novel oxygen-economical nano-photosensitizer via co-encapsulation of an OXPHOS inhibitor (ATO) and a newly developed type-I photosensitizer (IPS) into a polymeric micelle of PEG-b-PCL. By controlling the length of hydrophobic PCL segments, we successfully optimized the micelle size to around 30 nm for enhanced tumor penetration. The orchestration of the two functional components, ATO and IPS, can simultaneously hinder the two major tumor oxygen-consuming pathways, where ATO targets mitochondrial complex III to inhibit cellular respiration, while IPS generates ROS through a low oxygen-consuming type-I photochemical pathway, enabling remarkable PDT efficacies in both hypoxic cells and a 4T1 tumor-bearing BALB/c mouse model. This work sheds new light on the construction of nano-photosensitizers to rejuvenate PDT against hypoxic solid tumors.
Collapse
Affiliation(s)
- Hao Zhang
- School of Material Science and Engineering, Tiangong University, No. 399 BinShuiXi Road, Tianjin 300387, China.
| | - Xiaosa Yan
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| | - Yongkang Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| | - Chenlu Bao
- School of Material Science and Engineering, Tiangong University, No. 399 BinShuiXi Road, Tianjin 300387, China.
| | - Changhua Li
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| |
Collapse
|
48
|
Sivasubramanian M, Lo LW. Assessment of Nanoparticle-Mediated Tumor Oxygen Modulation by Photoacoustic Imaging. BIOSENSORS 2022; 12:336. [PMID: 35624636 PMCID: PMC9138624 DOI: 10.3390/bios12050336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 06/01/2023]
Abstract
Photoacoustic imaging (PAI) is an invaluable tool in biomedical imaging, as it provides anatomical and functional information in real time. Its ability to image at clinically relevant depths with high spatial resolution using endogenous tissues as contrast agents constitutes its major advantage. One of the most important applications of PAI is to quantify tissue oxygen saturation by measuring the differential absorption characteristics of oxy and deoxy Hb. Consequently, PAI can be utilized to monitor tumor-related hypoxia, which is a crucial factor in tumor microenvironments that has a strong influence on tumor invasiveness. Reactive oxygen species (ROS)-based therapies, such as photodynamic therapy, radiotherapy, and sonodynamic therapy, are oxygen-consuming, and tumor hypoxia is detrimental to their efficacy. Therefore, a persistent demand exists for agents that can supply oxygen to tumors for better ROS-based therapeutic outcomes. Among the various strategies, NP-mediated supplemental tumor oxygenation is especially encouraging due to its physio-chemical, tumor targeting, and theranostic properties. Here, we focus on NP-based tumor oxygenation, which includes NP as oxygen carriers and oxygen-generating strategies to alleviate hypoxia monitored by PAI. The information obtained from quantitative tumor oxygenation by PAI not only supports optimal therapeutic design but also serves as a highly effective tool to predict therapeutic outcomes.
Collapse
Affiliation(s)
| | - Leu-Wei Lo
- Department of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 350, Taiwan;
| |
Collapse
|
49
|
Lyu F, Li Y, Yan Z, He Q, Cheng L, Zhang P, Liu B, Liu C, Song Y, Xing Y. Identification of ISG15 and ZFP36 as novel hypoxia- and immune-related gene signatures contributing to a new perspective for the treatment of prostate cancer by bioinformatics and experimental verification. J Transl Med 2022; 20:202. [PMID: 35538543 PMCID: PMC9092714 DOI: 10.1186/s12967-022-03398-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022] Open
Abstract
Background Prostatic cancer (PCa) is one of the most common malignant tumors in men worldwide. Emerging evidence indicates significance of hypoxia and immunity in PCa invasion and metastasis. This study aimed to develop a hypoxia- and immune-related gene risk signature and explore the molecular mechanisms to formulate a better prognostic tool for PCa patients. Methods The hypoxia and immune scores of all PCa patients in The Cancer Genome Atlas (TCGA) dataset were calculated via the maximally selected rank statistics method and the ESTIMATE algorithm. From common genes identified overlapping hypoxia- and immune-related differentially expressed genes (DE-HRGs and DE-IRGs), a hypoxia- and immune-related gene risk signature was developed utilizing univariate and multivariate Cox regression analyses, and validated in the Memorial Sloan Kettering Cancer Centre (MSKCC) database. The immune cell infiltration level of PCa samples were evaluated with ssGSEA algorithm. Differential expression of prognostic genes was evidenced by immunohistochemistry and western blot (WB) in paired PCa samples. Expression levels of these genes and their variations under regular and hypoxic conditions were examined in cell lines. The functional effects of the prognostic gene on PCa cells were examined by wound healing and transwell assays. Results A hypoxia- and immune-related gene risk signature constructed by ISG15 and ZFP36 displays significant predictive potency, with higher risk score representing worse survival. A nomogram based on independent prognostic factors including the risk score and Gleason score exhibited excellent clinical value in the survival prediction of PCa. Infiltration levels of eosinophils, neutrophils, Tcm, Tem, TFH, Th1 cells, and Th17 cells were significantly lower in the high-risk group. Conversely, aDC, pDC, T helper cells, and Tregs were significantly higher. Additionally, the two prognostic genes were closely correlated with the tumor-infiltrating immune cell subset in PCa progression. RT-qPCR and WB presented higher and lower expression of ISG15 and ZFP36 in PCa cells, respectively. They were correspondingly increased and decreased in PCa cells under hypoxic conditions. Wound healing and transwell assays showed that over-expression of ISG15 promoted the migration and invasion of PCa cells. Conclusion Our study identified a novel hypoxia- and immune-related gene signature, contributing a new perspective to the treatment of PCa Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03398-4.
Collapse
Affiliation(s)
- Fang Lyu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yunxue Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhecheng Yan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qingliu He
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lulin Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Pu Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bing Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunyu Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yarong Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yifei Xing
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
50
|
Seledtsov VI, von Delwig AA. Oxygen therapy in traditional and immunotherapeutic treatment protocols of cancer patients: current reality and future prospects. Expert Rev Anticancer Ther 2022; 22:575-581. [PMID: 35468308 DOI: 10.1080/14737140.2022.2070153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The metabolic environment in ischemic and hypoxic tumors is known to contribute to cancer progression. Importantly, peculiar metabolic changes occurring in malignant cells (the increased glycolysis and the hampered Krebs cycle) may contribute to decreased antioxidant-dependent defense in ischemic and hypoxic tumors. AREAS COVERED In the clinic, oxygen saturation of tumors is usually achieved by the application of water-soluble ozone and hyperbaric oxygen therapy. Tumor oxygenation has been shown to inhibit tumor growth and potentiate anti-tumor effects of chemoradiotherapy in animal experiments and the clinical setting. Tumor oxygenation could enhance anti-tumor effects achieved by tumor blood vessel occlusion or angiostatic therapy. EXPERT OPINION Owing to a profound influence of ROS on both the innate and adaptive immunity, oxygen therapy, when combined simultaneously or sequentially with immunotherapeutic interventions (such as immune checkpoint inhibition, drug-induced immunostimulation, adoptive cell therapy, hyperthermia, etc.), could be considered as a novel highly-effective clinical biological approach to cancer treatment.
Collapse
Affiliation(s)
- Victor Ivanovich Seledtsov
- Department of Immunology, Innovita Research Company, Vilnius, Lithuania.,Center for Immunotherapy, Central Clinical Hospital of the Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|