1
|
Gong S, Yang L, Xu M, Xiang M, Lang J, Zhang H, Shan Y. A combined gene signature model for predicting radiotherapy response and relapse-free survival in laryngeal squamous cell carcinoma. Cancer Cell Int 2025; 25:102. [PMID: 40102978 PMCID: PMC11916850 DOI: 10.1186/s12935-025-03739-5] [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: 06/27/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Radioresistance is a major challenge in radiotherapy for laryngeal squamous cell carcinoma (LSCC), and there is currently no effective method to predict radiosensitivity in LSCC patients. This study aimed to establish a prediction model for radiotherapy response based on gene expression. METHODS The datasets of LSCC were obtained from the ENT department of Shanghai Ruijin Hospital and The Cancer Genome Atlas (TCGA). Lasso regression and Cox regression were used to establish the prediction model based on gene expression. Weighted gene coexpression network analysis (WGCNA) was used to analyze the correlation between gene expression and clinical characteristics. RT-qPCR was used to detect gene expression in tumor tissue to verify the accuracy of the prediction model. RESULTS Using a cohort of LSCC cases receiving radiotherapy collected in the TCGA database, the 3 protein-coding genes (PCGs) signature model was identified for the first time as the predictor of relapse-free survival and radiosensitivity in LSCC patients. And we explored the potential clinical value of 3 PCGs and screened out 2 long non-coding RNAs (lncRNAs) potential associated with 3 PCGs. More importantly, the LSCC cases collected by our department were used to preliminarily verify the predictive power of the 3 PCGs signature model for the radiosensitivity of LSCC, and the significant correlation between the expression levels of the 3 PCGs and the 2 lncRNAs. CONCLUSION We successfully establish a radiosensitivity prediction model based on the 3 PCGs Riskscore, which provides a theoretical basis for the decision-making of LSCC treatment options. Meantime, we preliminarily screen the potential associated lncRNAs of the 3 PCGs for further basic and clinical research.
Collapse
Affiliation(s)
- Shiqi Gong
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Liyun Yang
- Department of Otolaryngology, Gongli Hospital of Pudong, Shanghai, 200135, China
| | - Meng Xu
- Department of Oncology, Changhai Hospital Affiliated to Navy Medical University, Shanghai, 200433, China
| | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Juntian Lang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hao Zhang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yamin Shan
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
2
|
Rondeau JD, Lipari S, Mathieu B, Beckers C, Van de Velde JA, Mignion L, Da Silva Morais M, Kreuzer M, Colauzzi I, Capeloa T, Pruschy M, Gallez B, Sonveaux P. Mitochondria-targeted antioxidant MitoQ radiosensitizes tumors by decreasing mitochondrial oxygen consumption. Cell Death Discov 2024; 10:514. [PMID: 39730333 DOI: 10.1038/s41420-024-02277-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024] Open
Abstract
Hypoxic tumors are radioresistant stemming from the fact that oxygen promotes reactive oxygen species (ROS) propagation after water radiolysis and stabilizes irradiation-induced DNA damage. Therefore, an attractive strategy to radiosensitize solid tumors is to increase tumor oxygenation at the time of irradiation, ideally above a partial pressure of 10 mm-Hg at which full radiosensitization can be reached. Historically, the many attempts to increase vascular O2 delivery have had limited efficacy, but mathematical models predicted that inhibiting cancer cell respiration would be more effective. Here, we report that mitochondria-targeted antioxidant MitoQ can radiosensitize human breast tumors in mice. This was not a class effect, as neither MitoTEMPO nor SKQ1 shared this property. At clinically relevant nanomolar concentrations, MitoQ completely abrogated the oxygen consumption of several human cancer cell lines of different origins, which was associated with a glycolytic switch. Using orthotopic breast cancer models in mice, we observed that pretreating hypoxic MDA-MB-231 tumors with MitoQ delayed tumor growth with both single dose irradiation and clinically relevant fractionated radiotherapy. Oxygenated MCF7 tumors were not radiosensitized, suggesting an oxygen enhancement effect of MitoQ. Because MitoQ already successfully passed Phase I clinical trials, our findings foster its clinical evaluation in combination with radiotherapy.
Collapse
Affiliation(s)
- Justin D Rondeau
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Sara Lipari
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Barbara Mathieu
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium
| | - Claire Beckers
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Justine A Van de Velde
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Lionel Mignion
- Nuclear and Electron Spin Technologies (NEST) Platform, LDRI, UCLouvain, Brussels, Belgium
| | - Mauricio Da Silva Morais
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marvin Kreuzer
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ilaria Colauzzi
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Tania Capeloa
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium.
- WEL Research Institute, WELBIO Department, Wavre, Belgium.
| |
Collapse
|
3
|
Ćwiklińska A, Przewodowska D, Koziorowski D, Szlufik S. Innovative Approaches to Brain Cancer: The Use of Magnetic Resonance-guided Focused Ultrasound in Glioma Therapy. Cancers (Basel) 2024; 16:4235. [PMID: 39766134 PMCID: PMC11674718 DOI: 10.3390/cancers16244235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Gliomas are a wide group of common brain tumors, with the most aggressive type being glioblastoma multiforme (GBM), with a 5-year survival rate of less than 5% and a median survival time of approximately 12-14 months. The standard treatment of GBM includes surgical excision, radiotherapy, and chemotherapy with temozolomide (TMZ). However, tumor recurrence and progression are common. Therefore, more effective treatment for GBM should be found. One of the main obstacles to the treatment of GBM and other gliomas is the blood-brain barrier (BBB), which impedes the penetration of antitumor chemotherapeutic agents into glioblastoma cells. Nowadays, one of the most promising novel methods for glioma treatment is Magnetic Resonance-guided Focused Ultrasound (MRgFUS). Low-intensity FUS causes the BBB to open transiently, which allows better drug delivery to the brain tissue. Under magnetic resonance guidance, ultrasound waves can be precisely directed to the tumor area to prevent side effects in healthy tissues. Through the open BBB, we can deliver targeted chemotherapeutics, anti-tumor agents, immunotherapy, and gene therapy directly to gliomas. Other strategies for MRgFUS include radiosensitization, sonodynamic therapy, histotripsy, and thermal ablation. FUS can also be used to monitor the treatment and progression of gliomas using blood-based liquid biopsy. All these methods are still under preclinical or clinical trials and are described in this review to summarize current knowledge and ongoing trials.
Collapse
Affiliation(s)
| | | | | | - Stanisław Szlufik
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 03-242 Warsaw, Poland
| |
Collapse
|
4
|
Kumar P, Lacroix M, Dupré P, Arslan J, Fenou L, Orsetti B, Le Cam L, Racoceanu D, Radulescu O. Deciphering oxygen distribution and hypoxia profiles in the tumor microenvironment: a data-driven mechanistic modeling approach. Phys Med Biol 2024; 69:125023. [PMID: 38815610 DOI: 10.1088/1361-6560/ad524a] [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: 03/08/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
Objective. The distribution of hypoxia within tissues plays a critical role in tumor diagnosis and prognosis. Recognizing the significance of tumor oxygenation and hypoxia gradients, we introduce mathematical frameworks grounded in mechanistic modeling approaches for their quantitative assessment within a tumor microenvironment. By utilizing known blood vasculature, we aim to predict hypoxia levels across different tumor types.Approach. Our approach offers a computational method to measure and predict hypoxia using known blood vasculature. By formulating a reaction-diffusion model for oxygen distribution, we derive the corresponding hypoxia profile.Main results. The framework successfully replicates observed inter- and intra-tumor heterogeneity in experimentally obtained hypoxia profiles across various tumor types (breast, ovarian, pancreatic). Additionally, we propose a data-driven method to deduce partial differential equation models with spatially dependent parameters, which allows us to comprehend the variability of hypoxia profiles within tissues. The versatility of our framework lies in capturing diverse and dynamic behaviors of tumor oxygenation, as well as categorizing states of vascularization based on the dynamics of oxygen molecules, as identified by the model parameters.Significance. The proposed data-informed mechanistic method quantitatively assesses hypoxia in the tumor microenvironment by integrating diverse histopathological data and making predictions across different types of data. The framework provides valuable insights from both modeling and biological perspectives, advancing our comprehension of spatio-temporal dynamics of tumor oxygenation.
Collapse
Affiliation(s)
- P Kumar
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, INSERM, Montpellier, France
- Sorbonne Université, CNRS, INSERM, AP-HP, Inria, Paris Brain Institute (ICM), Paris, France
| | - M Lacroix
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - P Dupré
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - J Arslan
- Sorbonne Université, CNRS, INSERM, AP-HP, Inria, Paris Brain Institute (ICM), Paris, France
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye & Ear Hospital, East Melbourne, Australia
| | - L Fenou
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - B Orsetti
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - L Le Cam
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - D Racoceanu
- Sorbonne Université, CNRS, INSERM, AP-HP, Inria, Paris Brain Institute (ICM), Paris, France
| | - O Radulescu
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
5
|
Li CX, Gong ZC, Zhang WN, Zhang Y, Zhao HR. Radioresistance or/and radiosensitivity of head and neck squamous cell carcinoma: biological angle. Oral Maxillofac Surg 2024; 28:547-555. [PMID: 37935817 DOI: 10.1007/s10006-023-01189-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/29/2023] [Indexed: 11/09/2023]
Abstract
OBJECTIVE This narrative review aimed to compile and summarize clinically relevant literature in radiation therapy and to discuss the potential in radioresistant and radiosensitive head and neck squamous cell carcinoma (HNSCC). METHODS AND MATERIALS Google Scholar, PubMed, and the Cochrane Library were retrieved using combined key words such as "radiotherapy" and "head and neck cancer." Search strings additionally queried were "radioresistant," "radiosensitive," "head and neck region," "squamous cell carcinoma," in combination with Boolean operators 'AND' and 'OR.' Subsequently, the resulting publications were included for review of the full text. RESULTS Radiotherapeutic responses currently in clinical observation referred to HNSCC scoping were selected into this review. The compiled mechanisms were then detailed concerning on the clinical significance, biological characteristics, and molecular function. CONCLUSIONS Brachytherapy or/and external-beam radiotherapy are crucial for treating HNSCC especially the early stage patients, but in some patients with locally advanced tumors, their outcome with radiation therapy is poor due to obvious radioresistance. The curative effects mainly depend on the response to radiation therapy so an updated review is needed to optimize further applications in HNSCC radiotherapy.
Collapse
Affiliation(s)
- Chen-Xi Li
- Department of Oral and Maxillofacial Oncology & Surgery, School / Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, Urumqi, 830054, China.
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhong-Cheng Gong
- Department of Oral and Maxillofacial Oncology & Surgery, School / Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, Urumqi, 830054, China.
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
| | - Wei-Na Zhang
- Ear, Nose & Throat Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yang Zhang
- The First Ward of Oncological Department, Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Hua-Rong Zhao
- The First Ward of Oncological Department, Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| |
Collapse
|
6
|
Buyse C, Mignion L, Joudiou N, Melloul S, Driesschaert B, Gallez B. Sensitive simultaneous measurements of oxygenation and extracellular pH by EPR using a stable monophosphonated trityl radical and lithium phthalocyanine. Free Radic Biol Med 2024; 213:11-18. [PMID: 38218552 PMCID: PMC10923140 DOI: 10.1016/j.freeradbiomed.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/15/2024]
Abstract
The monitoring of acidosis and hypoxia is crucial because both factors promote cancer progression and impact the efficacy of anti-cancer treatments. A phosphonated tetrathiatriarylmethyl (pTAM) has been previously described to monitor both parameters simultaneously, but the sensitivity to tackle subtle changes in oxygenation was limited. Here, we describe an innovative approach combining the pTAM radical and lithium phthalocyanine (LiPc) crystals to provide sensitive simultaneous measurements of extracellular pH (pHe) and pO2. Both parameters can be measured simultaneously as both EPR spectra do not overlap, with a gain in sensitivity to pO2 variations by a factor of 10. This procedure was applied to characterize the impact of carbogen breathing in a breast cancer 4T1 model as a proof-of-concept. No significant change in pHe and pO2 was observed using pTAM alone, while LiPc detected a significant increase in tumor oxygenation. Interestingly, we observed that pTAM systematically overestimated the pO2 compared to LiPc. In addition, we analyzed the impact of an inhibitor (UK-5099) of the mitochondrial pyruvate carrier (MPC) on the tumor microenvironment. In vitro, the exposure of 4T1 cells to UK-5099 for 24 h induced a decrease in pHe and oxygen consumption rate (OCR). In vivo, a significant decrease in tumor pHe was observed in UK-5099-treated mice, while there was no change for mice treated with the vehicle. Despite the change observed in OCR, no significant change in tumor oxygenation was observed after the UK-5099 treatment. This approach is promising for assessing in vivo the effect of treatments targeting tumor metabolism.
Collapse
Affiliation(s)
- Chloe Buyse
- Biomedical Magnetic Resonance Research Group (REMA), Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium
| | - Lionel Mignion
- Nuclear and Electron Spin Technologies Platform (NEST), Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium
| | - Nicolas Joudiou
- Nuclear and Electron Spin Technologies Platform (NEST), Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium
| | - Samia Melloul
- Biomedical Magnetic Resonance Research Group (REMA), Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium
| | - Benoit Driesschaert
- Department of Pharmaceutical Sciences, School of Pharmacy & In Vivo Multifunctional Magnetic Resonance Center, West Virginia University, Morgantown, WV, USA
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group (REMA), Louvain Drug Research Institute (LDRI), UCLouvain, Brussels, Belgium.
| |
Collapse
|
7
|
Li CX, Tan XR, Wei W, Li MQ, Zhang WN, Gong ZC, Zhang Y, Zhao HR. A radiobiological perspective on radioresistance or/and radiosensitivity of head and neck squamous cell carcinoma. Rep Pract Oncol Radiother 2024; 28:809-822. [PMID: 38515813 PMCID: PMC10954264 DOI: 10.5603/rpor.99355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 03/23/2024] Open
Abstract
Background This article aimed to compile and summarize clinically relevant literature in radiation therapy, and to discuss the potential in radioresistant and radiosensitive head and neck cancer. Study Design Narrative review. Materials and methods Google Scholar, PubMed and the Cochrane Library were retrieved using combined key words such as "radiotherapy" and "head and neck cancer". Search strings additionally queried were "radioresistant", "radiosensitive", "head and neck region", "squamous cell carcinoma", in combination with Boolean Operators 'AND' and 'OR'. Subsequently, the resulting publications were included for review of the full text. Results Radiotherapeutic response currently in clinical observation referred to HNSCC scoping were selected into this review. The compiled mechanisms were then detailed concerning on the clinical significance, biological characteristics, and molecular function. Conclusions Brachytherapy or/and external-beam radiotherapy are crucial for treating HNSCC, especially the early stage patients, but in patients with locally advanced tumors, their outcome with radiation therapy is poor due to obvious radioresistance. The curative effects mainly depend on the response of radiation therapy, so an updated review is needed to optimize further applications in HNSCC radiotherapy.
Collapse
Affiliation(s)
- Chen-xi Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-rong Tan
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei Wei
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Mu-qiu Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei-na Zhang
- Ear, Nose & Throat Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhong-cheng Gong
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yang Zhang
- The First Ward of Oncological Department, Cancer Center, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hua-rong Zhao
- The First Ward of Oncological Department, Cancer Center, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
8
|
Tan X, Liao D, Rao C, Zhou L, Tan Z, Pan Y, Singh A, Kumar A, Liu J, Li B. Recent advances in nano-architectonics of metal-organic frameworks for chemodynamic therapy. J SOLID STATE CHEM 2022. [DOI: 10.1016/j.jssc.2022.123352] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
d’Hose D, Mathieu B, Mignion L, Hardy M, Ouari O, Jordan BF, Sonveaux P, Gallez B. EPR Investigations to Study the Impact of Mito-Metformin on the Mitochondrial Function of Prostate Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185872. [PMID: 36144606 PMCID: PMC9504708 DOI: 10.3390/molecules27185872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022]
Abstract
Background: Mito-metformin10 (MM10), synthesized by attaching a triphenylphosphonium cationic moiety via a 10-carbon aliphatic side chain to metformin, is a mitochondria-targeted analog of metformin that was recently demonstrated to alter mitochondrial function and proliferation in pancreatic ductal adenocarcinoma. Here, we hypothesized that this compound may decrease the oxygen consumption rate (OCR) in prostate cancer cells, increase the level of mitochondrial ROS, alleviate tumor hypoxia, and radiosensitize tumors. Methods: OCR and mitochondrial superoxide production were assessed by EPR (9 GHz) in vitro in PC-3 and DU-145 prostate cancer cells. Reduced and oxidized glutathione were assessed before and after MM10 exposure. Tumor oxygenation was measured in vivo using 1 GHz EPR oximetry in PC-3 tumor model. Tumors were irradiated at the time of maximal reoxygenation. Results: 24-hours exposure to MM10 significantly decreased the OCR of PC-3 and DU-145 cancer cells. An increase in mitochondrial superoxide levels was observed in PC-3 but not in DU-145 cancer cells, an observation consistent with the differences observed in glutathione levels in both cancer cell lines. In vivo, the tumor oxygenation significantly increased in the PC-3 model (daily injection of 2 mg/kg MM10) 48 and 72 h after initiation of the treatment. Despite the significant effect on tumor hypoxia, MM10 combined to irradiation did not increase the tumor growth delay compared to the irradiation alone. Conclusions: MM10 altered the OCR in prostate cancer cells. The effect of MM10 on the superoxide level was dependent on the antioxidant capacity of cell line. In vivo, MM10 alleviated tumor hypoxia, yet without consequence in terms of response to irradiation.
Collapse
Affiliation(s)
- Donatienne d’Hose
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Barbara Mathieu
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Lionel Mignion
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Micael Hardy
- Institut de Chimie Radicalaire UMR 7273, Aix-Marseille Université/CNRS, 13013 Marseille, France
| | - Olivier Ouari
- Institut de Chimie Radicalaire UMR 7273, Aix-Marseille Université/CNRS, 13013 Marseille, France
| | - Bénédicte F. Jordan
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherches Expérimentales et Cliniques (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Research Institute, 1300 Wavre, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Correspondence:
| |
Collapse
|
10
|
Kaeppler JR, Chen J, Buono M, Vermeer J, Kannan P, Cheng W, Voukantsis D, Thompson JM, Hill MA, Allen D, Gomes A, Kersemans V, Kinchesh P, Smart S, Buffa F, Nerlov C, Muschel RJ, Markelc B. Endothelial cell death after ionizing radiation does not impair vascular structure in mouse tumor models. EMBO Rep 2022; 23:e53221. [PMID: 35848459 PMCID: PMC9442312 DOI: 10.15252/embr.202153221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/15/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
The effect of radiation therapy on tumor vasculature has long been a subject of debate. Increased oxygenation and perfusion have been documented during radiation therapy. Conversely, apoptosis of endothelial cells in irradiated tumors has been proposed as a major contributor to tumor control. To examine these contradictions, we use multiphoton microscopy in two murine tumor models: MC38, a highly vascularized, and B16F10, a moderately vascularized model, grown in transgenic mice with tdTomato-labeled endothelium before and after a single (15 Gy) or fractionated (5 × 3 Gy) dose of radiation. Unexpectedly, even these high doses lead to little structural change of the perfused vasculature. Conversely, non-perfused vessels and blind ends are substantially impaired after radiation accompanied by apoptosis and reduced proliferation of their endothelium. RNAseq analysis of tumor endothelial cells confirms the modification of gene expression in apoptotic and cell cycle regulation pathways after irradiation. Therefore, we conclude that apoptosis of tumor endothelial cells after radiation does not impair vascular structure.
Collapse
Affiliation(s)
- Jakob R Kaeppler
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Jianzhou Chen
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Mario Buono
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Jenny Vermeer
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Pavitra Kannan
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Wei‐Chen Cheng
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Dimitrios Voukantsis
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - James M Thompson
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Mark A Hill
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Danny Allen
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Ana Gomes
- In Vivo ImagingThe Francis Crick InstituteLondonUK
| | - Veerle Kersemans
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Paul Kinchesh
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Sean Smart
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Francesca Buffa
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Claus Nerlov
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Ruth J Muschel
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
| | - Bostjan Markelc
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of OncologyUniversity of OxfordOxfordUK
- Present address:
Department of Experimental OncologyInstitute of Oncology LjubljanaLjubljanaSlovenia
| |
Collapse
|
11
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
12
|
Mudassar F, Shen H, Cook KM, Hau E. Improving the synergistic combination of programmed death‐1/programmed death ligand‐1 blockade and radiotherapy by targeting the hypoxic tumour microenvironment. J Med Imaging Radiat Oncol 2022; 66:560-574. [PMID: 35466515 PMCID: PMC9322583 DOI: 10.1111/1754-9485.13416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/05/2022] [Accepted: 04/10/2022] [Indexed: 11/28/2022]
Abstract
Immune checkpoint inhibition with PD‐1/PD‐L1 blockade is a promising area in the field of anti‐cancer therapy. Although clinical data have revealed success of PD‐1/PD‐L1 blockade as monotherapy or in combination with CTLA‐4 or chemotherapy, the combination with radiotherapy could further boost anti‐tumour immunity and enhance clinical outcomes due to the immunostimulatory effects of radiation. However, the synergistic combination of PD‐1/PD‐L1 blockade and radiotherapy can be challenged by the complex nature of the tumour microenvironment (TME), including the presence of tumour hypoxia. Hypoxia is a major barrier to the effectiveness of both radiotherapy and PD‐1/PD‐L1 blockade immunotherapy. Thus, targeting the hypoxic TME is an attractive strategy to enhance the efficacy of the combination. Addition of compounds that directly or indirectly reduce hypoxia, to the combination of PD‐1/PD‐L1 inhibitors and radiotherapy may optimize the success of the combination and improve therapeutic outcomes. In this review, we will discuss the synergistic combination of PD‐1/PD‐L1 blockade and radiotherapy and highlight the role of hypoxic TME in impeding the success of both therapies. In addition, we will address the potential approaches for targeting tumour hypoxia and how exploiting these strategies could benefit the combination of PD‐1/PD‐L1 blockade and radiotherapy.
Collapse
Affiliation(s)
- Faiqa Mudassar
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research The Westmead Institute for Medical Research Sydney New South Wales Australia
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
| | - Han Shen
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research The Westmead Institute for Medical Research Sydney New South Wales Australia
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
| | - Kristina M Cook
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Eric Hau
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research The Westmead Institute for Medical Research Sydney New South Wales Australia
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre Westmead Hospital Sydney New South Wales Australia
- Blacktown Hematology and Cancer Centre Blacktown Hospital Sydney New South Wales Australia
| |
Collapse
|
13
|
Ding Q, Xu Z, Zhou L, Rao C, Li W, Muddassir M, Sakiyama H, Li B, Ouyang Q, Liu J. A multimodal Metal-Organic framework based on unsaturated metal site for enhancing antitumor cytotoxicity through Chemo-Photodynamic therapy. J Colloid Interface Sci 2022; 621:180-194. [PMID: 35461133 DOI: 10.1016/j.jcis.2022.04.078] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023]
Abstract
Chemodynamic therapy when combined with chemotherapy opens up a new avenue for treatment of cancer. However, its development is still restricted by low targeting, high dose and toxic side effects. Herein, rational designing and construction of a new multifunctional platform with the core-shell structure 5-ALA@UiO-66-NH-FAM@CP1 (ALA = 5-aminolevulinic acid, CP1 = zirconium-pemetrexed (Zr-MTA)) has been performed. In this platform, CP1 acting as a shell is encapsulated with the UiO-66-NH2 to engender a core-shell structure that promotes and achieves a high MTA loading rate through high affinity between MTA and unsaturated Zr site of UiO-66-NH2. The 5-ALA and 5-carboxyl fluorescein (5-FAM) was successfully loaded and covalently combined with UiO-66-NH2 due to its high porosity and presence of amino groups. The characterization results indicated that the loading rate of MTA (41.03 wt%) of platform is higher than the reported values. More importantly, the in vitro and in vivo results also demonstrated that it has a good folate targeting ability and realizes high efficient antitumor activity by chemotherapy combied with photodynamic therapy (PDT). This newly developed multifunctional platform could provide a new idea for designing and constructing the carrier with chemotherapy and PDT therapy.
Collapse
Affiliation(s)
- Qiongjie Ding
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Luyi Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Congying Rao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Weimin Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mohd Muddassir
- Department of Chemistry, College of Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hiroshi Sakiyama
- Department of Science, Faculty of Science, Yamagata University, 1-4-12 Kojirakawa, Yamagata 990-8560, Japan
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Qin Ouyang
- Department of General Surgery, Dalang Hospital, Dongguan 523800, China.
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China.
| |
Collapse
|
14
|
Matsumoto KI, Nakanishi I, Zhelev Z, Bakalova R, Aoki I. Nitroxyl Radical as a Theranostic Contrast Agent in Magnetic Resonance Redox Imaging. Antioxid Redox Signal 2022; 36:95-121. [PMID: 34148403 PMCID: PMC8792502 DOI: 10.1089/ars.2021.0110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance:In vivo assessment of paramagnetic and diamagnetic conversions of nitroxyl radicals based on cyclic redox mechanism can be an index of tissue redox status. The redox mechanism of nitroxyl radicals, which enables their use as a normal tissue-selective radioprotector, is seen as being attractive on planning radiation therapy. Recent Advances:In vivo redox imaging using nitroxyl radicals as redox-sensitive contrast agents has been developed to assess tissue redox status. Chemical and biological behaviors depending on chemical structures of nitroxyl radical compounds have been understood in detail. Polymer types of nitroxyl radical contrast agents and/or nitroxyl radical-labeled drugs were designed for approaching theranostics. Critical Issues: Nitroxyl radicals as magnetic resonance imaging (MRI) contrast agents have several advantages compared with those used in electron paramagnetic resonance (EPR) imaging, while support by EPR spectroscopy is important to understand information from MRI. Redox-sensitive paramagnetic contrast agents having a medicinal benefit, that is, nitroxyl-labeled drug, have been developed and proposed. Future Directions: A development of suitable nitroxyl contrast agent for translational theranostic applications with high reaction specificity and low normal tissue toxicity is under progress. Nitroxyl radicals as redox-sensitive magnetic resonance contrast agents can be a useful tool to detect an abnormal tissue redox status such as disordered oxidative stress. Antioxid. Redox Signal. 36, 95-121.
Collapse
Affiliation(s)
- Ken-Ichiro Matsumoto
- Quantitative RedOx Sensing Group, Department of Radiation Regulatory Science Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Japan
| | - Ikuo Nakanishi
- Quantum RedOx Chemistry Group, Institute for Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Japan
| | - Zhivko Zhelev
- Medical Faculty, Trakia University, Stara Zagora, Bulgaria.,Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Rumiana Bakalova
- Functional and Molecular Imaging Goup, Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Japan
| | - Ichio Aoki
- Functional and Molecular Imaging Goup, Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Japan
| |
Collapse
|
15
|
Krysztofiak A, Szymonowicz K, Hlouschek J, Xiang K, Waterkamp C, Larafa S, Goetting I, Vega-Rubin-de-Celis S, Theiss C, Matschke V, Hoffmann D, Jendrossek V, Matschke J. Metabolism of cancer cells commonly responds to irradiation by a transient early mitochondrial shutdown. iScience 2021; 24:103366. [PMID: 34825138 PMCID: PMC8603201 DOI: 10.1016/j.isci.2021.103366] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 09/01/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022] Open
Abstract
Cancer bioenergetics fuel processes necessary to maintain viability and growth under stress conditions. We hypothesized that cancer metabolism supports the repair of radiation-induced DNA double-stranded breaks (DSBs). We combined the systematic collection of metabolic and radiobiological data from a panel of irradiated cancer cell lines with mathematical modeling and identified a common metabolic response with impact on the DSB repair kinetics, including a mitochondrial shutdown followed by compensatory glycolysis and resumption of mitochondrial function. Combining ionizing radiation (IR) with inhibitors of the compensatory glycolysis or mitochondrial respiratory chain slowed mitochondrial recovery and DNA repair kinetics, offering an opportunity for therapeutic intervention. Mathematical modeling allowed us to generate new hypotheses on general and individual mechanisms of the radiation response with relevance to DNA repair and on metabolic vulnerabilities induced by cancer radiotherapy. These discoveries will guide future mechanistic studies for the discovery of metabolic targets for overcoming intrinsic or therapy-induced radioresistance.
Collapse
Affiliation(s)
- Adam Krysztofiak
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Klaudia Szymonowicz
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Julian Hlouschek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Kexu Xiang
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Christoph Waterkamp
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, 45117 Essen, Germany
| | - Safa Larafa
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Isabell Goetting
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Silvia Vega-Rubin-de-Celis
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, 45117 Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
16
|
Ang MJY, Chan SY, Goh YY, Luo Z, Lau JW, Liu X. Emerging strategies in developing multifunctional nanomaterials for cancer nanotheranostics. Adv Drug Deliv Rev 2021; 178:113907. [PMID: 34371084 DOI: 10.1016/j.addr.2021.113907] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer involves a collection of diseases with a common trait - dysregulation in cell proliferation. At present, traditional therapeutic strategies against cancer have limitations in tackling various tumors in clinical settings. These include chemotherapeutic resistance and the inability to overcome intrinsic physiological barriers to drug delivery. Nanomaterials have presented promising strategies for tumor treatment in recent years. Nanotheranostics combine therapeutic and bioimaging functionalities at the single nanoparticle level and have experienced tremendous growth over the past few years. This review highlights recent developments of advanced nanomaterials and nanotheranostics in three main directions: stimulus-responsive nanomaterials, nanocarriers targeting the tumor microenvironment, and emerging nanomaterials that integrate with phototherapies and immunotherapies. We also discuss the cytotoxicity and outlook of next-generation nanomaterials towards clinical implementation.
Collapse
Affiliation(s)
- Melgious Jin Yan Ang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Siew Yin Chan
- Institute of Materials Research and Engineering, Agency for Science, Technology, and Research, Singapore 138634, Singapore
| | - Yi-Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jun Wei Lau
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore.
| |
Collapse
|
17
|
Oxygen-Sensitive MRI: A Predictive Imaging Biomarker for Tumor Radiation Response? Int J Radiat Oncol Biol Phys 2021; 110:1519-1529. [PMID: 33775857 DOI: 10.1016/j.ijrobp.2021.03.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE To develop a noninvasive prognostic imaging biomarker related to hypoxia to predict SABR tumor control. METHODS AND MATERIALS A total of 145 subcutaneous syngeneic Dunning prostate R3327-AT1 rat tumors were focally irradiated once using cone beam computed tomography guidance on a small animal irradiator at 225 kV. Various doses in the range of 0 to 100 Gy were administered, while rats breathed air or oxygen, and tumor control was assessed up to 200 days. Oxygen-sensitive magnetic resonance imaging (MRI) (T1-weighted, ΔR1, ΔR2*) was applied to 79 of these tumors at 4.7 T to assess response to an oxygen gas breathing challenge on the day before irradiation as a probe of tumor hypoxia. RESULTS Increasing radiation dose in the range of 0 to 90 Gy enhanced tumor control of air-breathing rats with a TCD50 estimated at 59.6 ± 1.5 Gy. Control was significantly improved at some doses when rats breathed oxygen during irradiation (eg, 40 Gy; P < .05), and overall there was a modest left shift in the control curve: TCD50(oxygen) = 53.1 ± 3.1 Gy (P < .05 vs air). Oxygen-sensitive MRI showed variable response to oxygen gas breathing challenge; the magnitude of T1-weighted signal response (%ΔSI) allowed stratification of tumors in terms of local control at 40 Gy. Tumors showing %ΔSI >0.922 with O2-gas breathing challenge showed significantly better control at 40 Gy during irradiation while breathing oxygen (75% vs 0%, P < .01). In addition, increased radiation dose (50 Gy) substantially overcame resistance, with 50% control for poorly oxygenated tumors. Stratification of dose-response curves based on %ΔSI >0.922 revealed different survival curves, with TCD50 = 36.2 ± 3.2 Gy for tumors responsive to oxygen gas breathing challenge; this was significantly less than the 54.7 ± 2.4 Gy for unresponsive tumors (P < .005), irrespective of the gas inhaled during tumor irradiation. CONCLUSIONS Oxygen-sensitive MRI allowed stratification of tumors in terms of local control at 40 Gy, indicating its use as a potential predictive imaging biomarker. Increasing dose to 50 Gy overcame radiation resistance attributable to hypoxia in 50% of tumors.
Collapse
|
18
|
A Mesoscale Computational Model for Microvascular Oxygen Transfer. Ann Biomed Eng 2021; 49:3356-3373. [PMID: 34184146 DOI: 10.1007/s10439-021-02807-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/01/2021] [Indexed: 01/06/2023]
Abstract
We address a mathematical model for oxygen transfer in the microcirculation. The model includes blood flow and hematocrit transport coupled with the interstitial flow, oxygen transport in the blood and the tissue, including capillary-tissue exchange effects. Moreover, the model is suited to handle arbitrarily complex vascular geometries. The purpose of this study is the validation of the model with respect to classical solutions and the further demonstration of its adequacy to describe the heterogeneity of oxygenation in the tissue microenvironment. Finally, we discuss the importance of these effects in the treatment of cancer using radiotherapy.
Collapse
|
19
|
Jiang M, Qin B, Luo L, Li X, Shi Y, Zhang J, Luo Z, Zhu C, Guan G, Du Y, You J. A clinically acceptable strategy for sensitizing anti-PD-1 treatment by hypoxia relief. J Control Release 2021; 335:408-419. [PMID: 34089792 DOI: 10.1016/j.jconrel.2021.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023]
Abstract
The hypoxic tumor microenvironment (TME) hinders the effectiveness of immunotherapy. Alleviating tumor hypoxia to improve the efficacy of immune checkpoint inhibitors (ICIs) represented by programmed cell death protein 1 (PD-1) antibody has become a meaningful strategy. In this study, we adopted three methods to alleviate hypoxia, including direct oxygen delivery using two different carriers and an indirect way involving HIF-1α inhibition. Both in vivo and in vitro experiments showed that liposomes modified with perfluorocarbon or hemoglobin (PFC@lipo or Hb@lipo) were able to efficiently load and release oxygen, relieving tumor hypoxia. However, the gas release behavior of PFC@lipo was uncontrollable, which might induce acute hyperoxia side effects during intravenous injection and reduce its biosafety. In contrast, whether administered locally or systemically, Hb@lipo revealed high animal tolerance, and was much safer than commercial HIF-1α inhibitor (PX-478), displaying prospects as a promising oxygen carrier for clinical practice. Pharmacodynamic experiments suggested that Hb@lipo helped PD-1 antibody break the therapeutic bottleneck and significantly inhibited the progression of 4 T1 breast cancer. But in CT26 colon cancer, the combination therapy failed to suppress tumor growth. After in-depth analysis and comparison, we found that the ratio of M1/M2 tumor associated macrophages (TAMs) between these two tumor models were dramatically different. And the lower M1/M2 ratio in CT26 tumors limited the anti-tumor effect of combination therapy. In this study, three methods for alleviating tumor hypoxia were compared from the perspectives of biosafety, efficacy and clinical applicability. Among them, Hb@lipo stood out, and its combined use with PD-1 antibody exhibit a distinct synergistic suppression effect on tumors with more M1 macrophages presented in the microenvironment. Our work provided a good reference for improving the efficacy of PD-1 antibody by alleviating tumor hypoxia.
Collapse
Affiliation(s)
- Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Bing Qin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Xiang Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Chunqi Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Guannan Guan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Yongzhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
20
|
Wu K, Chen X, Feng J, Zhang S, Xu Y, Zhang J, Wu Q, You M, Xia B, Ma S. Capilliposide C from Lysimachia capillipes Restores Radiosensitivity in Ionizing Radiation-Resistant Lung Cancer Cells Through Regulation of ERRFI1/EGFR/STAT3 Signaling Pathway. Front Oncol 2021; 11:644117. [PMID: 33869036 PMCID: PMC8047471 DOI: 10.3389/fonc.2021.644117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/12/2021] [Indexed: 12/25/2022] Open
Abstract
Aims Radiation therapy is used as the primary treatment for lung cancer. Unfortunately, radiation resistance remains to be the major clinic problem for lung cancer patients. Lysimachia capillipes capilliposide C (LC-C), an extract from LC Hemsl, has demonstrated multiple anti-cancer effects in several types of cancer. Here, we investigated the potential therapeutic impacts of LC-C on radiosensitivity in lung cancer cells and their underlying mechanisms. Methods Non-small cell lung cancer cell lines were initially irradiated to generate ionizing radiation (IR)-resistant lung cancer cell lines. RNA-seq analysis was used to examine the whole-transcriptome alteration in IR-resistant lung cancer cells treated with or without LC-C, and the differentially expressed genes with most significance were verified by RT-qPCR. Colony formation assays were performed to determine the effect of LC-C and the target gene ErbB receptor feedback inhibitor 1 (ERRFI1) on radiosensitivity of IR-resistant lung cancer cells. In addition, effects of ERRFI1 on cell cycle distribution, DNA damage repair activity were assessed by flow cytometry and γ-H2AX immunofluorescence staining respectively. Western blotting was performed to identify the activation of related signaling pathways. Tumor xenograft experiments were conducted to observe the effect of LC-C and ERRFI1 on radiosensitivity of IR-resistant lung cancer cells in vivo. Results Compared with parental cells, IR-resistant lung cancer cells were more resistant to radiation. LC-C significantly enhanced the effect of radiation in IR-resistant lung cancer cells both in vitro and in vivo and validated ERRFI1 as a candidate downstream gene by RNA-seq. Forced expression of ERRFI1 alone could significantly increase the radiosensitivity of IR-resistant lung cancer cells, while silencing of ERRFI1 attenuated the radiosensitizing function of LC-C. Accordingly, LC-C and ERRFI1 effectively inhibited IR-induced DNA damage repair, and ERRFI1 significantly induced G2/M checkpoint arrest. Additional investigations revealed that down-regulation of EGFR/STAT3 pathway played an important role in radiosensitization between ERRFI1 and LC-C. Furthermore, the high expression level of ERRFI1 was associated with high overall survival rates in lung cancer patients. Conclusions Treatment of LC-C may serve as a promising therapeutic strategy to overcome the radiation resistance and ERRFI1 may be a potential therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Kan Wu
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Xueqin Chen
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China.,Department of Thoracic Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Jianguo Feng
- Zhejiang Cancer Research Institute, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shirong Zhang
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Yasi Xu
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Jingjing Zhang
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Qiong Wu
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Mingliang You
- Hangzhou Cancer Institute, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Bing Xia
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China.,Department of Oncology, Jiande Second People's Hospital, Hangzhou, China
| | - Shenglin Ma
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
21
|
Matsumoto KI, Mitchell JB, Krishna MC. Multimodal Functional Imaging for Cancer/Tumor Microenvironments Based on MRI, EPRI, and PET. Molecules 2021; 26:1614. [PMID: 33799481 PMCID: PMC8002164 DOI: 10.3390/molecules26061614] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/23/2022] Open
Abstract
Radiation therapy is one of the main modalities to treat cancer/tumor. The response to radiation therapy, however, can be influenced by physiological and/or pathological conditions in the target tissues, especially by the low partial oxygen pressure and altered redox status in cancer/tumor tissues. Visualizing such cancer/tumor patho-physiological microenvironment would be a useful not only for planning radiotherapy but also to detect cancer/tumor in an earlier stage. Tumor hypoxia could be sensed by positron emission tomography (PET), electron paramagnetic resonance (EPR) oxygen mapping, and in vivo dynamic nuclear polarization (DNP) MRI. Tissue oxygenation could be visualized on a real-time basis by blood oxygen level dependent (BOLD) and/or tissue oxygen level dependent (TOLD) MRI signal. EPR imaging (EPRI) and/or T1-weighted MRI techniques can visualize tissue redox status non-invasively based on paramagnetic and diamagnetic conversions of nitroxyl radical contrast agent. 13C-DNP MRI can visualize glycometabolism of tumor/cancer tissues. Accurate co-registration of those multimodal images could make mechanisms of drug and/or relation of resulted biological effects clear. A multimodal instrument, such as PET-MRI, may have another possibility to link multiple functions. Functional imaging techniques individually developed to date have been converged on the concept of theranostics.
Collapse
Affiliation(s)
- Ken-ichiro Matsumoto
- Quantitative RedOx Sensing Group, Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Quantum Medical Science Directorate, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - James B. Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1002, USA;
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1002, USA;
| |
Collapse
|
22
|
Donatienne d'Hose, Danhier P, Northshield H, Isenborghs P, Jordan BF, Gallez B. A versatile EPR toolbox for the simultaneous measurement of oxygen consumption and superoxide production. Redox Biol 2020; 40:101852. [PMID: 33418140 PMCID: PMC7804984 DOI: 10.1016/j.redox.2020.101852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/30/2023] Open
Abstract
In this paper, we describe an assay to analyze simultaneously the oxygen consumption rate (OCR) and superoxide production in a biological system. The analytical set-up uses electron paramagnetic resonance (EPR) spectroscopy with two different isotopically-labelled sensors: 15N-PDT (4-oxo-2,2,6,6-tetramethylpiperidine-d16-15N-1-oxyl) as oxygen-sensing probe and 14N-CMH (1-hydroxy-3-methoxycarbonyl-2,2,5,5-tetramethylpyrrolidine, a cyclic hydroxylamine, as sensor of reactive oxygen species (ROS). The superoxide contribution to CMH oxidation is assessed using SOD or PEGSOD as controls. Because the EPR spectra are not superimposable, the variation of EPR linewidth of 15N-PDT (linked to OCR) and the formation of the nitroxide from 14N-CMH (linked to superoxide production) can be recorded simultaneously over time on a single preparation. The EPR toolbox was qualified in biological systems of increasing complexity. First, we used an enzymatic assay based on the hypoxanthine (HX)/xanthine oxidase (XO) which is a well described model of oxygen consumption and superoxide production. Second, we used a cellular model of superoxide production using macrophages exposed to phorbol 12-myristate 13-acetate (PMA) which stimulates the NADPH oxidase (NOX) to consume oxygen and produce superoxide. Finally, we exposed isolated mitochondria to established inhibitors of the electron transport chain (rotenone and metformin) in order to assess their impact on OCR and superoxide production. This EPR toolbox has the potential to screen the effect of intoxicants or drugs targeting the mitochondrial function. OCR and superoxide production are crucial to assess mitochondrial (dys)function. The EPR toolbox analyzes simultaneously the OCR and superoxide production. The EPR toolbox was validated in enzymatic system, cells and isolated mitochondria. The EPR toolbox has the potential to screen compounds altering mitochondrial function.
Collapse
Affiliation(s)
- Donatienne d'Hose
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Danhier
- Nuclear and Electron Spin Technologies (NEST) Platform, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Heidi Northshield
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pauline Isenborghs
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium.
| |
Collapse
|
23
|
Ali MY, Oliva CR, Noman ASM, Allen BG, Goswami PC, Zakharia Y, Monga V, Spitz DR, Buatti JM, Griguer CE. Radioresistance in Glioblastoma and the Development of Radiosensitizers. Cancers (Basel) 2020; 12:E2511. [PMID: 32899427 PMCID: PMC7564557 DOI: 10.3390/cancers12092511] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Ionizing radiation is a common and effective therapeutic option for the treatment of glioblastoma (GBM). Unfortunately, some GBMs are relatively radioresistant and patients have worse outcomes after radiation treatment. The mechanisms underlying intrinsic radioresistance in GBM has been rigorously investigated over the past several years, but the complex interaction of the cellular molecules and signaling pathways involved in radioresistance remains incompletely defined. A clinically effective radiosensitizer that overcomes radioresistance has yet to be identified. In this review, we discuss the current status of radiation treatment in GBM, including advances in imaging techniques that have facilitated more accurate diagnosis, and the identified mechanisms of GBM radioresistance. In addition, we provide a summary of the candidate GBM radiosensitizers being investigated, including an update of subjects enrolled in clinical trials. Overall, this review highlights the importance of understanding the mechanisms of GBM radioresistance to facilitate the development of effective radiosensitizers.
Collapse
Affiliation(s)
- Md Yousuf Ali
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52242, USA;
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.R.O.); (B.G.A.); (P.C.G.); (D.R.S.)
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Claudia R. Oliva
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.R.O.); (B.G.A.); (P.C.G.); (D.R.S.)
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Abu Shadat M. Noman
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong 4331, Bangladesh;
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Bryan G. Allen
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.R.O.); (B.G.A.); (P.C.G.); (D.R.S.)
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Prabhat C. Goswami
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.R.O.); (B.G.A.); (P.C.G.); (D.R.S.)
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Yousef Zakharia
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; (Y.Z.); (V.M.)
| | - Varun Monga
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; (Y.Z.); (V.M.)
| | - Douglas R. Spitz
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.R.O.); (B.G.A.); (P.C.G.); (D.R.S.)
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - John M. Buatti
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Corinne E. Griguer
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.R.O.); (B.G.A.); (P.C.G.); (D.R.S.)
- Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| |
Collapse
|
24
|
Swartz HM, Flood AB, Schaner PE, Halpern H, Williams BB, Pogue BW, Gallez B, Vaupel P. How best to interpret measures of levels of oxygen in tissues to make them effective clinical tools for care of patients with cancer and other oxygen-dependent pathologies. Physiol Rep 2020; 8:e14541. [PMID: 32786045 PMCID: PMC7422807 DOI: 10.14814/phy2.14541] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
It is well understood that the level of molecular oxygen (O2 ) in tissue is a very important factor impacting both physiology and pathological processes as well as responsiveness to some treatments. Data on O2 in tissue could be effectively utilized to enhance precision medicine. However, the nature of the data that can be obtained using existing clinically applicable techniques is often misunderstood, and this can confound the effective use of the information. Attempts to make clinical measurements of O2 in tissues will inevitably provide data that are aggregated over time and space and therefore will not fully represent the inherent heterogeneity of O2 in tissues. Additionally, the nature of existing techniques to measure O2 may result in uneven sampling of the volume of interest and therefore may not provide accurate information on the "average" O2 in the measured volume. By recognizing the potential limitations of the O2 measurements, one can focus on the important and useful information that can be obtained from these techniques. The most valuable clinical characterizations of oxygen are likely to be derived from a series of measurements that provide data about factors that can change levels of O2 , which then can be exploited both diagnostically and therapeutically. The clinical utility of such data ultimately needs to be verified by careful studies of outcomes related to the measured changes in levels of O2 .
Collapse
Affiliation(s)
- Harold M Swartz
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Ann Barry Flood
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
| | - Philip E Schaner
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Howard Halpern
- Department Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Benjamin B Williams
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Bernard Gallez
- Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Peter Vaupel
- Department Radiation Oncology, University Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Center Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
25
|
Waller J, Onderdonk B, Flood A, Swartz H, Shah J, Shah A, Aydogan B, Halpern H, Hasan Y. The clinical utility of imaging methods used to measure hypoxia in cervical cancer. Br J Radiol 2020; 93:20190640. [PMID: 32286849 PMCID: PMC7336054 DOI: 10.1259/bjr.20190640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 03/18/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
While it is well-established that hypoxia is a major factor that affects clinical outcomes in cervical cancer, widespread usage of clinically available methods to detect and evaluate hypoxia during the course of treatment have not been established. This review compares these methods, summarizes their strengths and weaknesses, and assesses the pathways for their useful employment to alter clinical practice. We conducted a search on PubMed for literature pertaining to imaging hypoxic cervical cancer, and implemented keywords related to oxygen measurement tools to improve the relevance of the search results.Oxygenation level-dependent applications of MRI have demonstrated hypoxia-induced radioresistance, and changes in cervix tumor oxygenation from hyperoxic therapy.The hypoxic areas within tumors can be indirectly identified in dynamic contrast-enhanced images, where they generally display low signal enhancement, and diffusion-weighted images, which demonstrates areas of restricted diffusion (which correlates with hypoxia). Positron emmision tomography, used independently and with other imaging modalities, has demonstrated utility in imaging hypoxia through tracers specific for low oxygen levels, like Cu-ATSM tracers and nitroimidazoles. Detecting hypoxia in the tumors of patients diagnosed with cervical cancer via medical imaging and non-imaging tools like electron paramagnetic resonance oximetry can be utilized clinically, such as for guiding radiation and post-treatment surveillance, for a more personalized approach to treatment. The merits of these methods warrant further investigation via comparative effectiveness research and large clinical trials into their clinical applications.
Collapse
Affiliation(s)
- Joseph Waller
- Drexel College of Medicine, 2900 W Queen Ln, PA 19129, United States
| | - Benjamin Onderdonk
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Ann Flood
- Department of Radiology, Dartmouth Geisel School of Medicine, 1 Rope Ferry Rd, NH 03755, United States
| | - Harold Swartz
- Department of Radiology, Dartmouth Geisel School of Medicine, 1 Rope Ferry Rd, NH 03755, United States
| | - Jaffer Shah
- Drexel College of Medicine, 2900 W Queen Ln, PA 19129, United States
| | - Asghar Shah
- Brown University, Providence, RI 02912, United States
| | - Bulent Aydogan
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Howard Halpern
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Yasmin Hasan
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| |
Collapse
|
26
|
Läsche M, Emons G, Gründker C. Shedding New Light on Cancer Metabolism: A Metabolic Tightrope Between Life and Death. Front Oncol 2020; 10:409. [PMID: 32300553 PMCID: PMC7145406 DOI: 10.3389/fonc.2020.00409] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Since the earliest findings of Otto Warburg, who discovered the first metabolic differences between lactate production of cancer cells and non-malignant tissues in the 1920s, much time has passed. He explained the increased lactate levels with dysfunctional mitochondria and aerobic glycolysis despite adequate oxygenation. Meanwhile, we came to know that mitochondria remain instead functional in cancer cells; hence, metabolic drift, rather than being linked to dysfunctional mitochondria, was found to be an active act of direct response of cancer cells to cell proliferation and survival signals. This metabolic drift begins with the use of sugars and the full oxidative phosphorylation via the mitochondrial respiratory chain to form CO2, and it then leads to the formation of lactic acid via partial oxidation. In addition to oncogene-driven metabolic reprogramming, the oncometabolites themselves alter cell signaling and are responsible for differentiation and metastasis of cancer cells. The aberrant metabolism is now considered a major characteristic of cancer within the past 15 years. However, the proliferating anabolic growth of a tumor and its spread to distal sites of the body is not explainable by altered glucose metabolism alone. Since a tumor consists of malignant cells and its tumor microenvironment, it was important for us to understand the bilateral interactions between the primary tumor and its microenvironment and the processes underlying its successful metastasis. We here describe the main metabolic pathways and their implications in tumor progression and metastasis. We also portray that metabolic flexibility determines the fate of the cancer cell and ultimately the patient. This flexibility must be taken into account when deciding on a therapy, since singular cancer therapies only shift the metabolism to a different alternative path and create resistance to the medication used. As with Otto Warburg in his days, we primarily focused on the metabolism of mitochondria when dealing with this scientific question.
Collapse
Affiliation(s)
- Matthias Läsche
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|
27
|
Arnold CR, Mangesius J, Skvortsova II, Ganswindt U. The Role of Cancer Stem Cells in Radiation Resistance. Front Oncol 2020; 10:164. [PMID: 32154167 PMCID: PMC7044409 DOI: 10.3389/fonc.2020.00164] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSC) are a distinct subpopulation within a tumor. They are able to self-renew and differentiate and possess a high capability to repair DNA damage, exhibit low levels of reactive oxygen species (ROS), and proliferate slowly. These features render CSC resistant to various therapies, including radiation therapy (RT). Eradication of all CSC is a requirement for an effective antineoplastic treatment and is therefore of utmost importance for the patient. This makes CSC the prime targets for any therapeutic approach. Albeit clinical data is still scarce, experimental data and first clinical trials give hope that CSC-targeted treatment has the potential to improve antineoplastic therapies, especially for tumors that are known to be treatment resistant, such as glioblastoma. In this review, we will discuss CSC in the context of RT, describe known mechanisms of resistance, examine the possibilities of CSC as biomarkers, and discuss possible new treatment approaches.
Collapse
Affiliation(s)
- Christoph Reinhold Arnold
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Mangesius
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ira-Ida Skvortsova
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria.,EXTRO-Lab, Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Ute Ganswindt
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Swartz HM, Vaupel P, Williams BB, Schaner PE, Gallez B, Schreiber W, Ali A, Flood AB. 'Oxygen Level in a Tissue' - What Do Available Measurements Really Report? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1232:145-153. [PMID: 31893405 DOI: 10.1007/978-3-030-34461-0_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the paper is to discuss what currently is feasible clinically to measure the level of oxygen and how that measurement can be clinically useful. Because oxygen in tissues is quite heterogeneous and all methods of measurement can only provide an average across heterogeneities at some spatial and temporal resolution, the values that are obtained may have limitations on their clinical utility. However, even if such limitations are significant, if one utilizes repeated measurements and focuses on changes in the measured levels, rather than 'absolute levels', it may be possible to obtain very useful clinical information. While these considerations are especially pertinent in cancer, they also pertain to most other types of pathology.
Collapse
Affiliation(s)
- H M Swartz
- Department Radiology, Dartmouth Medical School, Hanover, NH, USA. .,Section Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - P Vaupel
- Department Radiation Oncology, University Medical Center, Mainz, Germany
| | - B B Williams
- Department Radiology, Dartmouth Medical School, Hanover, NH, USA.,Section Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - P E Schaner
- Section Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - B Gallez
- Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - W Schreiber
- Department Radiology, Dartmouth Medical School, Hanover, NH, USA
| | - A Ali
- Department Radiation Oncology, Emory School of Medicine, Atlanta, GA, USA
| | - A B Flood
- Department Radiology, Dartmouth Medical School, Hanover, NH, USA
| |
Collapse
|
29
|
Coates JT, Skwarski M, Higgins GS. Targeting tumour hypoxia: shifting focus from oxygen supply to demand. Br J Radiol 2019; 92:20170843. [PMID: 29436847 PMCID: PMC6435066 DOI: 10.1259/bjr.20170843] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/01/2018] [Accepted: 02/06/2018] [Indexed: 11/05/2022] Open
Abstract
Tumour hypoxia is a well-recognised barrier to anti-cancer therapy and represents one of the best validated targets in oncology. Previous attempts to tackle hypoxia have focussed primarily on increasing tumour oxygen supply; however, clinical studies using this approach have yielded only modest clinical benefit, with often significant toxicity and practical limitations. Therefore, there are currently no anti-hypoxia treatments in widespread clinical use. As an emerging alternative strategy, we discuss the relevance of inhibiting tumour oxygen metabolism to alleviate hypoxia and highlight recently initiated clinical trials using this approach.
Collapse
Affiliation(s)
- James T Coates
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Michael Skwarski
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Geoff S Higgins
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Tran LBA, Cao-Pham TT, Jordan BF, Deschoemaeker S, Heyerick A, Gallez B. Impact of myo-inositol trispyrophosphate (ITPP) on tumour oxygenation and response to irradiation in rodent tumour models. J Cell Mol Med 2018; 23:1908-1916. [PMID: 30575283 PMCID: PMC6378184 DOI: 10.1111/jcmm.14092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 02/01/2023] Open
Abstract
Tumour hypoxia is a well-established factor of resistance in radiation therapy (RT). Myo-inositol trispyrophosphate (ITPP) is an allosteric effector that reduces the oxygen-binding affinity of haemoglobin and facilitates the release of oxygen by red blood cells. We investigated herein the oxygenation effect of ITPP in six tumour models and its radiosensitizing effect in two of these models. The evolution of tumour pO2 upon ITPP administration was monitored on six models using 1.2 GHz Electron Paramagnetic Resonance (EPR) oximetry. The effect of ITPP on tumour perfusion was assessed by Hoechst staining and the oxygen consumption rate (OCR) in vitro was measured using 9.5 GHz EPR. The therapeutic effect of ITPP with and without RT was evaluated on rhabdomyosarcoma and 9L-glioma rat models. ITPP enhanced tumour oxygenation in six models. The administration of 2 g/kg ITPP once daily for 2 days led to a tumour reoxygenation for at least 4 days. ITPP reduced the OCR in six cell lines but had no effect on tumour perfusion when tested on 9L-gliomas. ITPP plus RT did not improve the outcome in rhabdomyosarcomas. In 9L-gliomas, some of tumours receiving the combined treatment were cured while other tumours did not benefit from the treatment. ITPP increased oxygenation in six tumour models. A decrease in OCR could contribute to the decrease in tumour hypoxia. The association of RT with ITPP was beneficial for a few 9L-gliomas but was absent in the rhabdomyosarcomas.
Collapse
Affiliation(s)
- Ly-Binh-An Tran
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Thanh-Trang Cao-Pham
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | | - Bernard Gallez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
31
|
Mast JM, Kuppusamy P. Hyperoxygenation as a Therapeutic Supplement for Treatment of Triple Negative Breast Cancer. Front Oncol 2018; 8:527. [PMID: 30524959 PMCID: PMC6256245 DOI: 10.3389/fonc.2018.00527] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) refers to a group of biologically aggressive breast cancers that do not express estrogen, progesterone or epidermal growth factor receptor 2 hormone receptors. Each subset of TNBC has a unique molecular profile and may require specific treatments. A combination of surgery and chemotherapy followed by radiation therapy is the standard treatment mode for TNBC patients. Tumor oxygen status (hypoxia) is a key factor that may compromise the effectiveness of radiation treatment, as it is known that hypoxia can confer radiation resistance. In this study, we characterized MDA-MB-231 orthotropic xenograft tumors with respect to tumor oxygen level and their response to supplemental oxygen therapy in combination with paclitaxel and radiation therapy. We observed that the TNBC tumors became severely hypoxic (pO2 < 4 mmHg) within 1 week of tumor growth and responded poorly to administration of respiratory hyperoxygenation (100% O2) to mitigate hypoxia. However, periodic administration of supplemental oxygen (100% O2; 60 min/day for 21 days) showed a significant inhibitory effect on tumor volume when compared to control (1,023 ± 32 mm3 vs. 1,378 ± 114 mm3; p < 0.05). Combination of supplemental oxygen with paclitaxel and radiation therapy led to a significant reduction in tumor growth when compared to radiation alone (239 ± 40 mm3 vs. 390 ± 32 mm3; p < 0.05). The therapeutic enhancement by supplemental oxygen is possibly attributed to increase in tumor oxygenation with paclitaxel at the time of radiation treatment. These findings may have important implications in the understanding of the role of oxygen and supplemental oxygen therapy for the treatment of TNBC patients.
Collapse
Affiliation(s)
- Jesse M Mast
- Department of Radiology and Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Periannan Kuppusamy
- Department of Radiology and Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| |
Collapse
|
32
|
Zuo H, Tao J, Shi H, He J, Zhou Z, Zhang C. Platelet-mimicking nanoparticles co-loaded with W 18O 49 and metformin alleviate tumor hypoxia for enhanced photodynamic therapy and photothermal therapy. Acta Biomater 2018; 80:296-307. [PMID: 30223092 DOI: 10.1016/j.actbio.2018.09.017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/26/2018] [Accepted: 09/12/2018] [Indexed: 02/03/2023]
Abstract
W18O49-mediated photodynamic therapy (PDT) and photothermal therapy (PTT) are limited by the easily oxidized property and tumor hypoxia. Here, we report the development of platelet membranes as nanocarriers to co-load W18O49 nanoparticles (NPs) and metformin (PM-W18O49-Met NPs). Platelet membranes can protect W18O49 from oxidation and immune evasion, and increase the accumulation of W18O49 in tumor sites via the passive EPR effect and active adhesion between platelets and cancer cells. The introduction of metformin (Met), a typical anti-diabetic drug, can alleviate the tumor hypoxia through reducing oxygen consumption. As a result, ROS and heat generation are both greatly increased, as revealed by ROS/hypoxia imaging in vitro, IR thermal imaging in vivo and PET imaging in vivo. PM-W18O49-Met NPs show the improved therapeutic effects with greatly inhibited tumor growth and induced tumor cell apoptosis. Therefore, our work provides a novel strategy for simultaneous enhanced PDT and PTT, which is promising in bioapplication. STATEMENTE OF SIGNIFICANCE: W18O49-mediated photodynamic therapy and photothermal therapy are limited by the poor delivery of nanoparticles to tumors, the easily oxidized property, and tumor hypoxia environment, which will induce tumor treatment failure. Herein, we report the development of platelet membranes as nanocarriers to co-load W18O49 nanoparticles and metformin (PM-W18O49-Met NPs). Platelet membranes can protect W18O49 from oxidation and immune evasion, and increase the accumulation of W18O49 in tumor sites via the passive EPR effect and active adhesion. Metformin can alleviate the tumor hypoxia through reducing oxygen consumption. Hence, ROS and heat generation are both greatly increased. PM-W18O49-Met NPs show the improved therapeutic effects with greatly inhibited tumor growth and induced apoptosis. Therefore, our work provides a novel strategy in bioapplication.
Collapse
Affiliation(s)
- Huaqin Zuo
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu 210008, PR China; Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu 210008, PR China; Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225001, PR China
| | - Junxian Tao
- Department of Endocrinology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225001, PR China
| | - Hua Shi
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu 210008, PR China
| | - Jian He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu 210008, PR China.
| | - Zhengyang Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu 210008, PR China.
| | - Chao Zhang
- Collaborative Innovation Center of Chemistry for Life Sciences, College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, PR China.
| |
Collapse
|
33
|
Śniegocka M, Podgórska E, Płonka PM, Elas M, Romanowska-Dixon B, Szczygieł M, Żmijewski MA, Cichorek M, Markiewicz A, Brożyna AA, Słominski AT, Urbańska K. Transplantable Melanomas in Hamsters and Gerbils as Models for Human Melanoma. Sensitization in Melanoma Radiotherapy-From Animal Models to Clinical Trials. Int J Mol Sci 2018; 19:E1048. [PMID: 29614755 PMCID: PMC5979283 DOI: 10.3390/ijms19041048] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
The focus of the present review is to investigate the role of melanin in the radioprotection of melanoma and attempts to sensitize tumors to radiation by inhibiting melanogenesis. Early studies showed radical scavenging, oxygen consumption and adsorption as mechanisms of melanin radioprotection. Experimental models of melanoma in hamsters and in gerbils are described as well as their use in biochemical and radiobiological studies, including a spontaneously metastasizing ocular model. Some results from in vitro studies on the inhibition of melanogenesis are presented as well as radio-chelation therapy in experimental and clinical settings. In contrast to cutaneous melanoma, uveal melanoma is very successfully treated with radiation, both using photon and proton beams. We point out that the presence or lack of melanin pigmentation should be considered, when choosing therapeutic options, and that both the experimental and clinical data suggest that melanin could be a target for radiosensitizing melanoma cells to increase efficacy of radiotherapy against melanoma.
Collapse
Affiliation(s)
- Martyna Śniegocka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Ewa Podgórska
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Przemysław M Płonka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Martyna Elas
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Bożena Romanowska-Dixon
- Department of Ophthalmology and Ocular Oncology, Medical College of Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Małgorzata Szczygieł
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Michał A Żmijewski
- Department of Histology, Medical University of Gdansk, 80-210 Gdańsk, Poland.
| | - Mirosława Cichorek
- Department of Embryology, Medical University of Gdansk, 80-210 Gdańsk, Poland.
| | - Anna Markiewicz
- Department of Ophthalmology and Ocular Oncology, Medical College of Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| | - Anna A Brożyna
- Department of Tumor Pathology and Pathomorphology, Faculty of Health Sciences, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland.
- Department of Dermatology, Comprehensive Cancer Center Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Andrzej T Słominski
- Department of Dermatology, Comprehensive Cancer Center Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- VA Medical Center, Birmingham, AL 35294, USA.
| | - Krystyna Urbańska
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland.
| |
Collapse
|
34
|
Abstract
Glycolysis has long been considered as the major metabolic process for energy production and anabolic growth in cancer cells. Although such a view has been instrumental for the development of powerful imaging tools that are still used in the clinics, it is now clear that mitochondria play a key role in oncogenesis. Besides exerting central bioenergetic functions, mitochondria provide indeed building blocks for tumor anabolism, control redox and calcium homeostasis, participate in transcriptional regulation, and govern cell death. Thus, mitochondria constitute promising targets for the development of novel anticancer agents. However, tumors arise, progress, and respond to therapy in the context of an intimate crosstalk with the host immune system, and many immunological functions rely on intact mitochondrial metabolism. Here, we review the cancer cell-intrinsic and cell-extrinsic mechanisms through which mitochondria influence all steps of oncogenesis, with a focus on the therapeutic potential of targeting mitochondrial metabolism for cancer therapy.
Collapse
Affiliation(s)
- Paolo Ettore Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, 10124 Torino, Italy
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - José Manuel Bravo-San Pedro
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France
- Université Pierre et Marie Curie/Paris VI, 75006 Paris, France
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France
- INSERM, U1138, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France
- Université Pierre et Marie Curie/Paris VI, 75006 Paris, France
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France
- INSERM, U1138, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, 75015 Paris, France
- Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| |
Collapse
|
35
|
|
36
|
Amoedo ND, Obre E, Rossignol R. Drug discovery strategies in the field of tumor energy metabolism: Limitations by metabolic flexibility and metabolic resistance to chemotherapy. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:674-685. [PMID: 28213330 DOI: 10.1016/j.bbabio.2017.02.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/11/2017] [Accepted: 02/13/2017] [Indexed: 12/20/2022]
Abstract
The search for new drugs capable of blocking the metabolic vulnerabilities of human tumors has now entered the clinical evaluation stage, but several projects already failed in phase I or phase II. In particular, very promising in vitro studies could not be translated in vivo at preclinical stage and beyond. This was the case for most glycolysis inhibitors that demonstrated systemic toxicity. A more recent example is the inhibition of glutamine catabolism in lung adenocarcinoma that failed in vivo despite a strong addiction of several cancer cell lines to glutamine in vitro. Such contradictory findings raised several questions concerning the optimization of drug discovery strategies in the field of cancer metabolism. For instance, the cell culture models in 2D or 3D might already show strong limitations to mimic the tumor micro- and macro-environment. The microenvironment of tumors is composed of cancer cells of variegated metabolic profiles, supporting local metabolic exchanges and symbiosis, but also of immune cells and stroma that further interact with and reshape cancer cell metabolism. The macroenvironment includes the different tissues of the organism, capable of exchanging signals and fueling the tumor 'a distance'. Moreover, most metabolic targets were identified from their increased expression in tumor transcriptomic studies, or from targeted analyses looking at the metabolic impact of particular oncogenes or tumor suppressors on selected metabolic pathways. Still, very few targets were identified from in vivo analyses of tumor metabolism in patients because such studies are difficult and adequate imaging methods are only currently being developed for that purpose. For instance, perfusion of patients with [13C]-glucose allows deciphering the metabolomics of tumors and opens a new area in the search for effective targets. Metabolic imaging with positron emission tomography and other techniques that do not involve [13C] can also be used to evaluate tumor metabolism and to follow the efficiency of a treatment at a preclinical or clinical stage. Relevant descriptors of tumor metabolism are now required to better stratify patients for the development of personalized metabolic medicine. In this review, we discuss the current limitations in basic research and drug discovery in the field of cancer metabolism to foster the need for more clinically relevant target identification and validation. We discuss the design of adapted drug screening assays and compound efficacy evaluation methods for the discovery of innovative anti-cancer therapeutic approaches at the level of tumor energetics. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- N D Amoedo
- University of Bordeaux, U1211MRGM, Bordeaux, France; INSERM, U1211MRGM, Bordeaux, France; Instituto de Bioquímica Médica Leopoldo De Meis, UFRJ, Rio de Janeiro, Brazil
| | - E Obre
- University of Bordeaux, U1211MRGM, Bordeaux, France; INSERM, U1211MRGM, Bordeaux, France; CELLOMET, Bordeaux, France
| | - R Rossignol
- University of Bordeaux, U1211MRGM, Bordeaux, France; INSERM, U1211MRGM, Bordeaux, France; CELLOMET, Bordeaux, France.
| |
Collapse
|
37
|
|