51
|
Jin K, He M, Chen B, Zhou X, Zhang C, Zhang Z, Hu D, Jiang Z, Wei Q, Qiu S, Yang L. A single-sample mRNA molecular classification of bladder cancer predicting prognosis and response to immunotherapy. Transl Androl Urol 2022; 11:943-958. [PMID: 35958899 PMCID: PMC9360513 DOI: 10.21037/tau-21-887] [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: 10/05/2021] [Accepted: 05/18/2022] [Indexed: 11/12/2022] Open
Abstract
Background As an immunogenic cancer, crosstalk between cancer cells and immune cells has been gradually recognized in bladder cancer (BC). Several studies have emphasized the clinical significance of the molecular stratification of BC without highlighting the role of the immune microenvironment. Although immunotherapy acted as a prospective treatment, more precise molecular stratification should be established to select those sensitive to immunotherapy. Methods To select specific immune genes forming subtypes indicating disparate prognoses, we performed bioinformatic analysis using BC transcriptomic profiles from six published datasets, with 408 BC samples in The Cancer Genome Atlas (TCGA) database and 295 individuals in International Cancer Genome Consortium (ICGC) database. Survival analyses were conducted using Kaplan-Meier curves, while Kruskal-Wallis tests were applied to test the differences among groups. Except for unsupervised clustering based on the differential expression of genes, we additionally performed binomial logistic regression, focusing on the mRNA level of a single sample. Results Unsupervised clustering showed that 4 clusters captured the best segmentation. After validation with survival data and simplification using binomial logistic regression, we found that cluster B and cluster D showed worse survival outcomes (P=0.012). Considering the similar survival outcomes of these two clusters, we recombined and performed another survival analysis, which also showed significant survival differences (P=0.0041). Bonding with clinical data, a greater proportion of risk factors were assigned to the worse prognosis subtype, especially showing higher grades in the subtype (P<0.001). In addition, immune cell infiltration, single nucleotide polymorphism (SNP) and copy number variation (CNV) all showed differences between clusters, indicating changes in the immune microenvironment and mutation burden. Through phenotypical analysis, we found metabolism and proliferation phenotypes associated with the immune clusters and mutually exclusive in BC, of which proliferation contributed to worse outcomes. Using the tumor immune dysfunction and exclusion (TIDE) score, a worse immunotherapy benefit was predicted in clusters B&D, defined as the worse prognosis subtype. Conclusions With this novel clustering criterion based on immune-related genes, we provide a better understanding of the immune microenvironment, further guiding the use of immunotherapy.
Collapse
Affiliation(s)
- Kun Jin
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Mingjing He
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Chen
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chichen Zhang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zilong Zhang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Hu
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongyuan Jiang
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shi Qiu
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Center of Biomedical Big Data, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Yang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
52
|
Maxwell AWP, Mendoza HG, Sellitti MJ, Camacho JC, Deipolyi AR, Ziv E, Sofocleous CT, Yarmohammadi H, Maybody M, Humm JL, Schwartz J, Juluru K, Dunphy MP, Boas FE. Optimizing 90Y Particle Density Improves Outcomes After Radioembolization. Cardiovasc Intervent Radiol 2022; 45:958-969. [PMID: 35459960 PMCID: PMC10103908 DOI: 10.1007/s00270-022-03139-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/28/2022] [Indexed: 01/19/2023]
Abstract
PURPOSE To determine how particle density affects dose distribution and outcomes after lobar radioembolization. METHODS Matched pairs of patients, treated with glass versus resin microspheres, were selected by propensity score matching (114 patients), in this single-institution retrospective study. For each patient, tumor and liver particle density (particles/cm3) and dose (Gy) were determined. Tumor-to-normal ratio was measured on both 99mTc-MAA SPECT/CT and post-90Y bremsstrahlung SPECT/CT. Microdosimetry simulations were used to calculate first percentile dose, which is the dose in the cold spots between microspheres. Local progression-free survival (LPFS) and overall survival were analyzed. RESULTS As more particles were delivered, doses on 90Y SPECT/CT became more uniform throughout the treatment volume: tumor and liver doses became more similar (p = 0.04), and microscopic cold spots between particles disappeared. For hypervascular tumors (tumor-to-normal ratio ≥ 2.6 on MAA scan), delivering fewer particles (< 6000 particles/cm3 treatment volume) was associated with better LPFS (p = 0.03). For less vascular tumors (tumor-to-normal ratio < 2.6), delivering more particles (≥ 6000 particles/cm3) was associated with better LPFS (p = 0.02). In matched pairs of patients, using the optimal particle density resulted in improved overall survival (11.5 vs. 6.8 months, p = 0.047), compared to using suboptimal particle density. Microdosimetry resulted in better predictions of LPFS (p = 0.03), and overall survival (p = 0.02), compared to conventional dosimetry. CONCLUSION The number of particles delivered can be chosen to maximize the tumor dose and minimize the liver dose, based on tumor vascularity. Optimizing the particle density resulted in improved LPFS and overall survival.
Collapse
Affiliation(s)
- Aaron W P Maxwell
- Department of Diagnostic Imaging, The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Humberto G Mendoza
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew J Sellitti
- Radiology Informatics, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan C Camacho
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amy R Deipolyi
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Etay Ziv
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Constantinos T Sofocleous
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hooman Yarmohammadi
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Majid Maybody
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jazmin Schwartz
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Krishna Juluru
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark P Dunphy
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - F Edward Boas
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
53
|
Griazov A, Griazov A, Grydina N, Stuley V. Stereotactic radiosurgery of radioresistant glioblastomas. The ways of overcoming radioresistance of hypoxic tumors. УКРАЇНСЬКИЙ РАДІОЛОГІЧНИЙ ТА ОНКОЛОГІЧНИЙ ЖУРНАЛ 2022. [DOI: 10.46879/ukroj.2.2022.25-40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Background. Taking into account high degree of resistance of glioblastoma to radiation therapy, and also low overall survival rates of patients, it is necessary to develop improved methods of treating this pathology, in particular, complex combined treatment with radiation therapy and radiosensitizers.
Purpose – to assess the effectiveness of radiosensitization of hypoxic tumors in radiosurgical treatment of glioblastomas; to increase non-recurrent and overall survival rate of patients. Materials and methods. Stereotactic radiosurgery (SRS) of glioblastoma was performed in 106 patients (average age – 53 years), 66 males (62,26%) and 40 females (37,73%). The average dose was 18 Gy in a single-fraction SRS, and 32 Gy (7 Gy per fraction) in multi-fraction SRS. The average volume tumor was 29 cm3 . The treatment group consisted of 66 patients who underwent SRS with radiosensitization. 40 patients made up the control group and underwent SRS without radiosensitization.
Results. Median overall survival (MOS) was 20 months in the group with radiosensitization, whereas in the control group it was 12 months. 10-month recurrence-free period after radiosurgery was observed in 95,4% of the patients of the group with radiosensitization and in 70,6% of the patients of the control group. MOS after SRS was similar between the patients with wild-type IDH tumors and patients with tumors with IDH mutation (10,0 months and 11,0 months respectively), and also between the patients with MGMT-methylated tumors and patients with MGMT-nonmethylated tumors (11,2 and 10,2 months respectively). Among all the treated patients, in 20 of them (16,6%) side radiation effects after SRS were observed, and in 9 patients (7,5%) radiation necrosis developed in 3 to 16 months after SRS. The signs of moderate toxicity in the form of vomiting were observed in 6,6% of the patients of the subgroup with metronidazole. There were no signs of toxicity in the subgroup with nimorazole.
Conclusions. Radiosensitization improves rates of overall survival by 53,3% and recurrence-free survival by 24,8 % in performing SRS of hypoxic radioresistant glioblastomas. Nimorazole and metronidazole are powerful radiosensitizers which increase radiosensitivity of tumor cells through enhancing oxygen saturation of hypoxic cells. In order to determine indications for performing SRS with radiosensitization and periods for performing an SRS session we must take into consideration the result of an oxygen test (level of oxygen saturation of the tumor), the peak of signal intensity in the zone of active tumor growth and the peak of saturation of the whole tumor volume.
Collapse
|
54
|
Wang Y, Lang J, Zuo JZ, Dong Y, Hu Z, Xu X, Zhang Y, Wang Q, Yang L, Wong STC, Wang H, Li H. The radiomic-clinical model using the SHAP method for assessing the treatment response of whole-brain radiotherapy: a multicentric study. Eur Radiol 2022; 32:8737-8747. [PMID: 35678859 DOI: 10.1007/s00330-022-08887-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To develop and validate a pretreatment magnetic resonance imaging (MRI)-based radiomic-clinical model to assess the treatment response of whole-brain radiotherapy (WBRT) by using SHapley Additive exPlanations (SHAP), which is derived from game theory, and can explain the output of different machine learning models. METHODS We retrospectively enrolled 228 patients with brain metastases from two medical centers (184 in the training cohort and 44 in the validation cohort). Treatment responses of patients were categorized as a non-responding group vs. a responding group according to the Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) criteria. For each tumor, 960 features were extracted from the MRI sequence. The least absolute shrinkage and selection operator (LASSO) was used for feature selection. A support vector machine (SVM) model incorporating clinical factors and radiomic features wase used to construct the radiomic-clinical model. SHAP method explained the SVM model by prioritizing the importance of features, in terms of assessment contribution. RESULTS Three radiomic features and three clinical factors were identified to build the model. Radiomic-clinical model yielded AUCs of 0.928 (95%CI 0.901-0.949) and 0.851 (95%CI 0.816-0.886) for assessing the treatment response in the training cohort and validation cohort, respectively. SHAP summary plot illustrated the feature's value affected the feature's impact attributed to model, and SHAP force plot showed the integration of features' impact attributed to individual response. CONCLUSION The radiomic-clinical model with the SHAP method can be useful for assessing the treatment response of WBRT and may assist clinicians in directing personalized WBRT strategies in an understandable manner. KEY POINTS • Radiomic-clinical model can be useful for assessing the treatment response of WBRT. • SHAP could explain and visualize radiomic-clinical machine learning model in a clinician-friendly way.
Collapse
Affiliation(s)
- Yixin Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China.,Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Jinwei Lang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Joey Zhaoyu Zuo
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Yaqin Dong
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Zongtao Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Xiuli Xu
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Yongkang Zhang
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Qinjie Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Lizhuang Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China.,Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX, 77030, USA
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China. .,University of Science and Technology of China, Hefei, 230026, People's Republic of China. .,Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| | - Hai Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China. .,University of Science and Technology of China, Hefei, 230026, People's Republic of China. .,Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| |
Collapse
|
55
|
Interplay between Caveolin-1 and body and tumor size affects clinical outcomes in breast cancer. Transl Oncol 2022; 22:101464. [PMID: 35660849 PMCID: PMC9166433 DOI: 10.1016/j.tranon.2022.101464] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Caveolin-1 (CAV1) is associated with cholesterol-rich membrane raft domains and is a master regulator of cell signaling and membrane transport. Here, we investigated CAV1's role in cellular compartments of breast cancer in relation to signaling pathways, clinicopathological features, and clinical outcomes. METHODS CAV1 levels were evaluated with immunohistochemistry in cytoplasm of invasive tumor cells and stromal cells in tumor tissue microarrays from a cohort of 1018 breast cancer patients (inclusion 2002-2012, Sweden). Cytoplasmic and stromal CAV1 were categorized as positive/negative and strong/not strong, respectively. CAV1 expression in relation to clinical outcomes was assessed with Cox regression. Investigations into CAV1 functional pathways was conducted in the STRING, GOBO, and TCGA databases. RESULTS CAV1 expression was associated with non-luminal subtypes, cell cycle control, inflammation, epithelial-mesenchymal transition, and the IGF/Insulin system. Generally, CAV1 was not associated with recurrence risk. Stromal CAV1's impact on recurrence risk was modified by BMI ≥25 kg/m2 (Pinteraction = 0.002), waist ≥80 cm (Pinteraction = 0.005), and invasive tumor size (pT2/3/4) (Pinteraction = 0.028). In low-risk patients only, strong stromal CAV1 significantly increased recurrence risk (HRsadj ≥1.61). In all patients, positive cytoplasmic CAV1 conferred >2-fold risk for contralateral disease HRadj 2.63 (95% CI 1.36-5.10). Strong stromal CAV1 conferred nearly 2-fold risk for locoregional recurrence HRadj 1.88 (95% CI 1.09-3.24). CONCLUSIONS CAV1's prognostic impact depended on its localization, anthropometric, and tumor factors. Stromal CAV1 predicted high recurrence risk in a group of supposedly 'low-risk' patients. Cytoplasmic CAV1 predicted metachronous contralateral disease. If confirmed, CAV1 could be used as treatment target and for risk-stratification.
Collapse
|
56
|
Xu AL, Xue YY, Tao WT, Wang SQ, Xu HQ. Oleanolic acid combined with olaparib enhances radiosensitization in triple negative breast cancer and hypoxia imaging with 18F-FETNIM micro PET/CT. Biomed Pharmacother 2022; 150:113007. [PMID: 35483190 DOI: 10.1016/j.biopha.2022.113007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
The heterogeneity of triple negative breast cancer (TNBC) results in the worst prognosis among breast cancer types, making its treatment strategy very challenging. A recent study showed that oleanolic acid (OA) has a radiosensitizing effect on tumor cells, but it does not show a good clinical effect when used alone in radiotherapy. The cytotoxicity of radiotherapy can be enhanced by modulating DNA repair, so new treatment options are being investigated to inhibit DNA repair pathways and sensitize tumors to radiation. Radiation induces DNA double-strand breaks (DSBs), and inhibition of Poly (ADP-Ribose) polymerase (PARP) can prevent the repair of these lesions. Hence, we evaluated the radiosensitization and the underlying mechanism of combination treatment with OA and olaparib in TNBC. Meanwhile, tumor hypoxia was monitored with 18F-Fluoroerythronitroimidazole (FETNIM) positron emission tomography/computed tomography (PET/CT) during radiosensitization therapy. Here, we found that OA and olaparib in combination with radiotherapy significantly inhibited cell proliferation compared with other groups. The results were observed using colony formation assays [sensitization enhancement ratios (SER) 1.16-1.65]. In vivo, tumor growth was significantly delayed in transplanted tumors receiving irradiation (IR) with OA and olaparib. 18F-FETNIM PET/CT can be utilized for tumor hypoxia monitoring and radiosensitization response evaluation. In conclusion, these results suggest that the combination of OA and olaparib with IR enhances the inhibition of MDA-MB-231 in cell culture and in mice, providing a potentially novel combination for the effective treatment of TNBC patients.
Collapse
Affiliation(s)
- A-Lei Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yang-Yang Xue
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wei-Tao Tao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Si-Qi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hui-Qin Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
57
|
Gao M, Huang X, Wu Z, Wang L, Yuan S, Du Z, Luo S, Li R, Wang W. Synthesis of a versatile mitochondria-targeting small molecule for cancer near-infrared fluorescent imaging and radio/photodynamic/photothermal synergistic therapies. Mater Today Bio 2022; 15:100316. [PMID: 35721281 PMCID: PMC9198388 DOI: 10.1016/j.mtbio.2022.100316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 12/17/2022]
Abstract
Although as a mainstay modal for cancer treatment, the clinical effect of radiotherapy (RT) does not yet meet the need of cancer patients. Developing tumour-preferential radiosensitizers or combining RT with other treatments has been acknowledged highly necessary to enhance the efficacy of RT. The present study reported a multifunctional bioactive small-molecule (designated as IR-83) simultaneously exhibiting tumour-preferential accumulation, near-infrared imaging and radio/photodynamic/photothermal therapeutic effects. IR-83 was designed and synthesized by introducing 2-nitroimidazole as a radiosensitizer into the framework of heptamethine cyanine dyes inherently with tumour-targeting and photosensitizing effects. As results, IR-83 preferentially accumulated in tumours, suppressed tumour growth and metastasis by integrating radio/photodynamic/photothermal multimodal therapies. Mechanism studies showed that IR-83 accumulated in cancer cell mitochondria, induced excessive reactive oxygen species (ROS), and generated high heat after laser irradiation. On one hand, these phenomena led to mitochondrial dysfunction and a sharp decline in oxidative phosphorylation to lessen tissue oxygen consumption. On the other hand, excessive ROS in mitochondria destroyed the balance of antioxidants and oxidative stress balance by down-regulating the intracellular antioxidant system, and subsequently sensitized ionizing radiation-generated irreversible DNA double-strand breaks. Therefore, this study presented a promising radiosensitizer and a new alternative strategy to enhance RT efficacy via mitochondria-targeting multimodal synergistic treatment. IR-83 is chemically synthesized via introduction of a radiosensitizing moiety into a cancer-targeting heptamethine cyanine framework.. IR-83 exhibits multifunctional bioactivities of cancer-preferential accumulation, near infrared imaging-guided multimodal treatment. IR-83 exerts a synergistic therapeutic effect of RT/PDT/PTT by targeting cancer cell mitochondria. Cancer radiotherapy is significantly sensitized by mitochondria-targeting delivery of a radiosensitizing moiety, PTT-triggered increase of O2 level and PDT-induced irreversible DNA double-strand breaks.
Collapse
Affiliation(s)
- Mingquan Gao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Xie Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zifei Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shaolong Yuan
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zaizhi Du
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shenglin Luo
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Rong Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Corresponding author. No. 30, Gaotanyan Zheng Street, Shapingba District, Chongqing, China.
| | - Weidong Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
- Corresponding author. No. 55, section 4, Renmin South Road, Chengdu, Sichuan Province, China.
| |
Collapse
|
58
|
Depletion of HIF-1α by Inducible Cre/loxP Increases the Sensitivity of Cultured Murine Hepatocytes to Ionizing Radiation in Hypoxia. Cells 2022; 11:cells11101671. [PMID: 35626708 PMCID: PMC9139307 DOI: 10.3390/cells11101671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023] Open
Abstract
The transcription factor hypoxia-inducible factor (HIF) is the main oxygen sensor which regulates adaptation to cellular hypoxia. The aim of this study was to establish cultured murine hepatocyte derived cells (mHDC) as an in vitro model and to analyze the role of HIF-1α in apoptosis induction, DNA damage repair and sensitivity to ionizing radiation (IR). We have crossed C57/BL6 mice that bear loxP sites flanking exon 2 of Hif1a with mice which carry tamoxifen-inducible global Cre expression. From the offspring, we have established transduced hepatocyte cultures which are permanently HIF-1α deficient after tamoxifen treatment. We demonstrated that the cells produce albumin, acetylcholine esterase, and the cytokeratins 8 and 18 which functionally characterizes them as hepatocytes. In moderate hypoxia, HIF-1α deficiency increased IR-induced apoptosis and significantly reduced the surviving fraction of mHDC as compared to HIF-1α expressing cells in colony formation assays. Furthermore, HIF-1α knockout cells displayed increased IR-induced DNA damage as demonstrated by increased generation and persistence of γH2AX foci. HIF-1α deficient cells showed delayed DNA repair after IR in hypoxia in neutral comet assays which may indicate that non-homologous end joining (NHEJ) repair capacity was affected. Overall, our data suggest that HIF-1α inactivation increases radiation sensitivity of mHDC cells.
Collapse
|
59
|
Feng B, Wu J, Shen B, Jiang F, Feng J. Cancer-associated fibroblasts and resistance to anticancer therapies: status, mechanisms, and countermeasures. Cancer Cell Int 2022; 22:166. [PMID: 35488263 PMCID: PMC9052457 DOI: 10.1186/s12935-022-02599-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment (TME) with diverse functions such as extracellular matrix (ECM) remodeling, modulation of metabolism and angiogenesis, and crosstalk with both cancer cells and infiltrating immune cells by production of growth factors, cytokines, and chemokines. Within the TME milieu, CAFs exhibit morphological and functional transitions with relatively specific markers and hold tremendous potential to facilitate tumorigenesis, development, and resistance towards multiple therapeutic strategies including chemotherapy, radiotherapy, targeted therapy, anti-angiogenesis therapy, immunotherapy, and endocrine therapy. Accordingly, CAFs themselves and the downstream effectors and/or signaling pathways are potential targets for optimizing the sensitivity of anti-cancer therapies. This review aims to provide a detailed landscape of the role that CAFs play in conferring therapeutic resistance in different cancers and the underlying mechanisms. The translational and therapeutic perspectives of CAFs in the individualized treatment of malignant tumors are also discussed.
Collapse
Affiliation(s)
- Bing Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Jianzhong Wu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Bo Shen
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Feng Jiang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China.
| | - Jifeng Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China.
| |
Collapse
|
60
|
He X, Lee B, Jiang Y. Extracellular matrix in cancer progression and therapy. MEDICAL REVIEW (2021) 2022; 2:125-139. [PMID: 37724245 PMCID: PMC10471113 DOI: 10.1515/mr-2021-0028] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/31/2022] [Indexed: 09/20/2023]
Abstract
The tumor ecosystem with heterogeneous cellular compositions and the tumor microenvironment has increasingly become the focus of cancer research in recent years. The extracellular matrix (ECM), the major component of the tumor microenvironment, and its interactions with the tumor cells and stromal cells have also enjoyed tremendously increased attention. Like the other components of the tumor microenvironment, the ECM in solid tumors differs significantly from that in normal organs and tissues. We review recent studies of the complex roles the tumor ECM plays in cancer progression, from tumor initiation, growth to angiogenesis and invasion. We highlight that the biomolecular, biophysical, and mechanochemical interactions between the ECM and cells not only regulate the steps of cancer progression, but also affect the efficacy of systemic cancer treatment. We further discuss the strategies to target and modify the tumor ECM to improve cancer therapy.
Collapse
Affiliation(s)
- Xiuxiu He
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Byoungkoo Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
61
|
Lin J, Chen S, Xiao L, Wang Z, Lin Y, Xu S. Identifying Molecular Subtypes and 6-Gene Prognostic Signature Based on Hypoxia for Optimizing Targeted Therapies in Non-Small Cell Lung Cancer. Int J Gen Med 2022; 15:4417-4432. [PMID: 35509605 PMCID: PMC9058021 DOI: 10.2147/ijgm.s352238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/09/2022] [Indexed: 11/23/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) accounts for a great number of all lung cancer cases. Hypoxia, one of the hallmarks in solid cancer, is closely involved in cancer cell progression and migration. This study aimed to develop a molecular subtyping system based on hypoxia-related genes and construct a prognostic model for NSCLC patients. Methods Unsupervised consensus clustering was used to classify molecular subtypes. Mutation and immune analyses were conducted to compare differences among the molecular subtypes. Univariate Cox regression, least absolute shrinkage and selection operator (LASSO) analysis, and step Akaike information criterion (stepAIC) were performed to screen prognostic genes. Results Two molecular subtypes (C1 and C2) were identified based on hypoxia-related genes and showed significant differences in survival, enriched pathways, tumor microenvironment (TME), and sensitivity to immunotherapy and chemotherapy. Interestingly, C1 subtype had better survival and response to targeted therapies. Oncogenic pathways, such as hypoxia, epithelial mesenchymal transition (EMT), NOTCH signaling, and p53 signaling pathways were more enriched in C2 subtype. A 6-gene prognostic model with robust ability was developed to classify NSCLC patients into high-risk and low-risk groups. Conclusion The novel molecular subtypes could assist personalized therapies to select suitable patients. The six prognostic genes may be novel targets for further understanding mechanisms of NSCLC development associated with hypoxia and exploiting novel targeted therapies.
Collapse
Affiliation(s)
- Jingrong Lin
- Department of Respiratory Medicine, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China
| | - Shujiao Chen
- Academic Affairs Office, People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China
| | - Linling Xiao
- The First Clinical Medical College, Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China
| | - Ziyan Wang
- Department of Respiratory Medicine, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China
| | - Yanqing Lin
- Department of Respiratory Medicine, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China
| | - Shungui Xu
- Department of Respiratory Medicine, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China
- Correspondence: Shungui Xu, Department of Respiratory Medicine, Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fujian, 35003, People’s Republic of China, Email
| |
Collapse
|
62
|
Du Z, Liu H, Bai L, Yan D, Li H, Peng S, Cao J, Liu SB, Tang Z. A Radiosensitivity Prediction Model Developed Based on Weighted Correlation Network Analysis of Hypoxia Genes for Lower-Grade Glioma. Front Oncol 2022; 12:757686. [PMID: 35280808 PMCID: PMC8916576 DOI: 10.3389/fonc.2022.757686] [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: 08/12/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background and PurposeHypoxia is one of the basic characteristics of the physical microenvironment of solid tumors. The relationship between radiotherapy and hypoxia is complex. However, there is no radiosensitivity prediction model based on hypoxia genes. We attempted to construct a radiosensitivity prediction model developed based on hypoxia genes for lower-grade glioma (LGG) by using weighted correlation network analysis (WGCNA) and least absolute shrinkage and selection operator (Lasso).MethodsIn this research, radiotherapy-related module genes were selected after WGCNA. Then, Lasso was performed to select genes in patients who received radiotherapy. Finally, 12 genes (AGK, ETV4, PARD6A, PTP4A2, RIOK3, SIGMAR1, SLC34A2, SMURF1, STK33, TCEAL1, TFPI, and UROS) were included in the model. A radiosensitivity-related risk score model was established based on the overall rate of The Cancer Genome Atlas (TCGA) dataset in patients who received radiotherapy. The model was validated in TCGA dataset and two Chinese Glioma Genome Atlas (CGGA) datasets. A novel nomogram was developed to predict the overall survival of LGG patients.ResultsWe developed and verified a radiosensitivity-related risk score model based on hypoxia genes. The radiosensitivity-related risk score served as an independent prognostic indicator. This radiosensitivity-related risk score model has prognostic prediction ability. Moreover, a nomogram integrating risk score with age and tumor grade was established to perform better for predicting 1-, 3-, and 5-year survival rates.ConclusionsWe developed and validated a radiosensitivity prediction model that can be used by clinicians and researchers to predict patient survival rates and achieve personalized treatment of LGG.
Collapse
Affiliation(s)
- Zixuan Du
- Department of Biostatistics and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China
| | - Hanshan Liu
- Department of Medical Oncology, Jiangsu Provincial Corps Hospital, Chinese People’s Armed Police Forces, Yangzhou City, China
| | - Lu Bai
- Department of Biostatistics and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China
| | - Derui Yan
- Department of Biostatistics and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Huijun Li
- Department of Biostatistics and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Sun Peng
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - JianPing Cao
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Song-Bai Liu
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China
- *Correspondence: Zaixiang Tang, ; Song-Bai Liu,
| | - Zaixiang Tang
- Department of Biostatistics and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
- *Correspondence: Zaixiang Tang, ; Song-Bai Liu,
| |
Collapse
|
63
|
Papaioannou L, Avgoustakis K. Responsive nanomedicines enhanced by or enhancing physical modalities to treat solid cancer tumors: Preclinical and clinical evidence of safety and efficacy. Adv Drug Deliv Rev 2022; 181:114075. [PMID: 34883140 DOI: 10.1016/j.addr.2021.114075] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/22/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022]
Abstract
Nanomedicine has improved cancer treatment but not to the extent anticipated. Responsive nanomedicines enhanced by physical modalities (radiation, ultrasounds, alternating magnetic fields) or enhancing the activity of physical modalities such as radiotherapy to kill cancer represents an important approach in improving the safety and anticancer effectiveness. Importantly, the combined treatments have shown promise for the treatment of difficult to treat tumors, such as tumors that are resistant to chemotherapy (multi drug resistant, MDR) or radiotherapy and hypoxic tumors, and for the prevention of tumor metastasis. In this review, the mechanisms of responsive nanomedicines activity enhancement by physical means and vice versa are presented and preclinical and, most importantly, clinical evidence of the safety and efficacy of nanomedicines enhanced by or enhancing by physical modalities in treating solid tumors are critically discussed.
Collapse
Affiliation(s)
- Ligeri Papaioannou
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Konstantinos Avgoustakis
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, 26504 Patras, Greece; Biomedical Research Foundation Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, Athens 11527, Greece.
| |
Collapse
|
64
|
Alsner J, Overgaard J, Tramm T, Lindegaard JC. Hypoxic gene expression is a prognostic factor for disease free survival in a cohort of locally advanced squamous cell cancer of the uterine cervix. Acta Oncol 2022; 61:172-178. [PMID: 34586924 DOI: 10.1080/0284186x.2021.1979249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Tumour hypoxia in locally advanced squamous cervical cancer (LACC) has been shown to be of substantial prognostic importance. The aims of the present study were therefore to investigate if hypoxia could be identified by a newly validated hypoxic gene expression classifier and used as a prognostic factor for disease free survival (DFS). MATERIAL AND METHODS Paraffin embedded biopsies were obtained from 190 patients with LACC with squamous cell carcinoma treated 2005-2016 with chemo-radiation and image guided adaptive brachytherapy. Analysis of hypoxia was successful in 183 patients (96%). Hypoxic classification of tumours into 'more' or 'less' hypoxic was based on 15 genes using the same method as in a prospective head and neck cancer trial (NCT02661152). HPV was genotyped using INNO-LiPA. Local tumour invasion was evaluated by the T-score. Primary endpoint was DFS analysed by Kaplan-Meier and Cox regression. Events were death of any cause, persistent disease, or recurrence. RESULTS The FIGO2009 stage distribution was IB-IIA 9%, IIB 64%, and III-IVA 27%, and mean T-score was 7.2. Pathological nodes were present in 53%. Median observation time was 5.2 years. Local control rate at 5 years was 96%, and pelvic (loco-regional) control 91%. Overall, 36% of the tumours were classified as 'more' hypoxic. The frequency of 'more' hypoxic tumours increased with local tumour intrusion (30% for T-score 0-9 vs. 55% for T-score ≥10, p = 0.004). Hypoxia was associated with decreased DFS in univariate, HR 1.71 (1.04-2.82), and multivariate analysis, HR 1.75 (1.04-2.92), and the effect was particularly observed among tumours with a T-score ≥10. HPV 16/18 was not associated with improved DFS in neither in univariate nor in multivariate analysis. CONCLUSION Hypoxic gene expression is a prominent prognostic factor for DFS in LACC with SCC histology and should be considered for treatment stratification in clinical trials.
Collapse
Affiliation(s)
- Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
65
|
Low-Dose-Rate Irradiation Suppresses the Expression of Cell Cycle-Related Genes, Resulting in Modification of Sensitivity to Anti-Cancer Drugs. Cells 2022; 11:cells11030501. [PMID: 35159310 PMCID: PMC8833988 DOI: 10.3390/cells11030501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 02/06/2023] Open
Abstract
The biological effects of low-dose-rate (LDR) radiation exposure in nuclear power plant accidents and medical uses of ionizing radiation (IR), although being a social concern, remain unclear. In this study, we evaluated the effects of LDR-IR on global gene expression in human cells and aimed to clarify the mechanisms. RNA-seq analyses demonstrated that relatively low dose rates of IR modify gene expression levels in TIG-3 cells under normoxic conditions, but those effects were attenuated under hypoxia-mimicking conditions. Gene set enrichment analysis demonstrated that LDR-IR significantly decreased gene expression related to cell division, cell cycle, mitosis, and the Aurora kinase B and FOXM1 pathways. Quantitative RT-PCR confirmed the down-regulation of AURKB and FOXM1 genes in TIG-3 cells with LDR-IR or hypoxia-mimicking treatments without any dose-rate effect. Knock-down experiments suggested that HIF-1α and HIF-2α, as well as DEC1, participated in down-regulation of AURKB and FOXM1 under DFOM treatments, but to a lesser extent under LDR-IR treatment. FACS and microscopic analyses demonstrated that LDR-IR induced G0/G1 arrest and increased micronucleus or chromosome condensation. Finally, MTT assays demonstrated that LDR-IR decreased sensitivity to paclitaxel or barasertib in TIG-3 cells but not in A549 cells. In conclusion, LDR-IR modifies global gene expression and cell cycle control, resulting in a reduction of sensitivity to anti-cancer chemotherapy in non-cancer cells and thus a reduction in untoward effects (GA).
Collapse
|
66
|
Scherschinski L, Prem M, Kremenetskaia I, Tinhofer I, Vajkoczy P, Karbe AG, Onken JS. Regulation of the Receptor Tyrosine Kinase AXL in Response to Therapy and Its Role in Therapy Resistance in Glioblastoma. Int J Mol Sci 2022; 23:ijms23020982. [PMID: 35055167 PMCID: PMC8781963 DOI: 10.3390/ijms23020982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
The receptor tyrosine kinase AXL (RTK-AXL) is implicated in therapy resistance and tumor progression in glioblastoma multiforme (GBM). Here, we investigated therapy-induced receptor modifications and how endogenous RTK-AXL expression and RTK-AXL inhibition contribute to therapy resistance in GBM. GBM cell lines U118MG and SF126 were exposed to temozolomide (TMZ) and radiation (RTX). Receptor modifications in response to therapy were investigated on protein and mRNA levels. TMZ-resistant and RTK-AXL overexpressing cell lines were exposed to increasing doses of TMZ and RTX, with and without RTK-AXL tyrosine kinase inhibitor (TKI). Colorimetric microtiter (MTT) assay and colony formation assay (CFA) were used to assess cell viability. Results showed that the RTK-AXL shedding product, C-terminal AXL (CT-AXL), rises in response to repeated TMZ doses and under hypoxia, acts as a surrogate marker for radio-resistance. Endogenous RTX-AXL overexpression leads to therapy resistance, whereas combination therapy of TZM and RTX with TKI R428 significantly increases therapeutic effects. This data proves the role of RTK-AXL in acquired and intrinsic therapy resistance. By demonstrating that therapy resistance may be overcome by combining AXL TKI with standard treatments, we have provided a rationale for future study designs investigating AXL TKIs in GBM.
Collapse
Affiliation(s)
- Lea Scherschinski
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Markus Prem
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
- Department of Neurosurgery, Technische Universität Dresden, 01069 Dresden, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany;
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung–DKTK), Partner Site Berlin, 10115 Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Anna-Gila Karbe
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Julia Sophie Onken
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung–DKTK), Partner Site Berlin, 10115 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)30-450-660253
| |
Collapse
|
67
|
Zhang X, He C, Xiang G. Engineering nanomedicines to inhibit hypoxia-inducible Factor-1 for cancer therapy. Cancer Lett 2022; 530:110-127. [PMID: 35041892 DOI: 10.1016/j.canlet.2022.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/18/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1), an essential promoter of tumor progression, has attracted increasing attention as a therapeutic target. In addition to hypoxic cellular conditions, HIF-1 activation can be triggered by cancer treatment, which causes drug tolerance and therapeutic failure. To date, a series of effective strategies have been explored to suppress HIF-1 function, including silencing the HIF-1α gene, inhibiting HIF-1α protein translation, degrading HIF-1α protein, and inhibiting HIF-1 transcription. Furthermore, nanoparticle-based drug delivery systems have been widely developed to improve the stability and pharmacokinetics of HIF-1 inhibitors or achieve HIF-1-targeted combination therapies as a nanoplatform. In this review, we summarize the current literature on nanomedicines targeting HIF-1 to combat cancer and discuss their potential for future development.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuanchuan He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
68
|
Feng N, Liu Y, Dai X, Wang Y, Guo Q, Li Q. Advanced applications of cerium oxide based nanozymes in cancer. RSC Adv 2022; 12:1486-1493. [PMID: 35425183 PMCID: PMC8979138 DOI: 10.1039/d1ra05407d] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cerium oxide nanozymes have emerged as a new type of bio-antioxidants in recent years. CeO2 nanozymes possess enzyme mimetic activities with outstanding free radical scavenging activity, facile synthesis conditions, and excellent biocompatibility. Based on these extraordinary properties, use of CeO2 nanozymes has been demonstrated to be a highly versatile therapeutic method for many diseases, such as for inflammation, rheumatoid arthritis, hepatic ischemia-reperfusion injury and Alzheimer's disease. In addition to that, CeO2 nanozymes have been widely used in the diagnosis and treatment of cancer. Many examples can be found in the literature, such as magnetic resonance detection, tumour marker detection, chemotherapy, radiotherapy, photodynamic therapy (PDT), and photothermal therapy (PTT). This review systematically summarises the latest applications of CeO2-based nanozymes in cancer research and treatment. We believe that this paper will help develop value-added CeO2 nanozymes, offering great potential in the biotechnology industry and with great significance for the diagnosis and treatment of a wide range of malignancies.
Collapse
Affiliation(s)
- Na Feng
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University Zhengzhou 450001 China
| | - Ying Liu
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University Zhengzhou 450001 China
| | - Xianglin Dai
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University Zhengzhou 450001 China
| | - Yingying Wang
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University Zhengzhou 450001 China
| | - Qiong Guo
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University Zhengzhou 450001 China
| | - Qing Li
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University Zhengzhou 450001 China
| |
Collapse
|
69
|
Brender JR, Saida Y, Devasahayam N, Krishna MC, Kishimoto S. Hypoxia Imaging As a Guide for Hypoxia-Modulated and Hypoxia-Activated Therapy. Antioxid Redox Signal 2022; 36:144-159. [PMID: 34428981 PMCID: PMC8856011 DOI: 10.1089/ars.2021.0176] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Significance: Oxygen imaging techniques, which can probe the spatiotemporal heterogeneity of tumor oxygenation, could be of significant clinical utility in radiation treatment planning and in evaluating the effectiveness of hypoxia-activated prodrugs. To fulfill these goals, oxygen imaging techniques should be noninvasive, quantitative, and capable of serial imaging, as well as having sufficient temporal resolution to detect the dynamics of tumor oxygenation to distinguish regions of chronic and acute hypoxia. Recent Advances: No current technique meets all these requirements, although all have strengths in certain areas. The current status of positron emission tomography (PET)-based hypoxia imaging, oxygen-enhanced magnetic resonance imaging (MRI), 19F MRI, and electron paramagnetic resonance (EPR) oximetry are reviewed along with their strengths and weaknesses for planning hypoxia-guided, intensity-modulated radiation therapy and detecting treatment response for hypoxia-targeted prodrugs. Critical Issues: Spatial and temporal resolution emerges as a major concern for these areas along with specificity and quantitative response. Although multiple oxygen imaging techniques have reached the investigative stage, clinical trials to test the therapeutic effectiveness of hypoxia imaging have been limited. Future Directions: Imaging elements of the redox environment besides oxygen by EPR and hyperpolarized MRI may have a significant impact on our understanding of the basic biology of the reactive oxygen species response and may extend treatment possibilities.
Collapse
Affiliation(s)
- Jeffrey R. Brender
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
70
|
Qin S, Xu Y, Li H, Chen H, Yuan Z. Recent advances in in situ oxygen-generating and oxygen-replenishing strategies for hypoxic-enhanced photodynamic therapy. Biomater Sci 2021; 10:51-84. [PMID: 34882762 DOI: 10.1039/d1bm00317h] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer is a leading cause of death worldwide, accounting for an estimated 10 million deaths by 2020. Over the decades, various strategies for tumor therapy have been developed and evaluated. Photodynamic therapy (PDT) has attracted increasing attention due to its unique characteristics, including low systemic toxicity and minimally invasive nature. Despite the excellent clinical promise of PDT, hypoxia is still the Achilles' heel associated with its oxygen-dependent nature related to increased tumor proliferation, angiogenesis, and distant metastases. Moreover, PDT-mediated oxygen consumption further exacerbates the hypoxia condition, which will eventually lead to the poor effect of drug treatment and resistance and irreversible tumor metastasis, even limiting its effective application in the treatment of hypoxic tumors. Hypoxia, with increased oxygen consumption, may occur in acute and chronic hypoxia conditions in developing tumors. Tumor cells farther away from the capillaries have much lower oxygen levels than cells in adjacent areas. However, it is difficult to change the tumor's deep hypoxia state through different ways to reduce the tumor tissue's oxygen consumption. Therefore, it will become more difficult to cure malignant tumors completely. In recent years, numerous investigations have focused on improving PDT therapy's efficacy by providing molecular oxygen directly or indirectly to tumor tissues. In this review, different molecular oxygen supplementation methods are summarized to alleviate tumor hypoxia from the innovative perspective of using supplemental oxygen. Besides, the existing problems, future prospects and potential challenges of this strategy are also discussed.
Collapse
Affiliation(s)
- Shuheng Qin
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Yue Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Hua Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| |
Collapse
|
71
|
Keulers TG, Koch A, van Gisbergen MW, Barbeau LMO, Zonneveld MI, de Jong MC, Savelkouls KGM, Wanders RG, Bussink J, Melotte V, Rouschop KMA. ATG12 deficiency results in intracellular glutamine depletion, abrogation of tumor hypoxia and a favorable prognosis in cancer. Autophagy 2021; 18:1898-1914. [PMID: 34904929 PMCID: PMC9450974 DOI: 10.1080/15548627.2021.2008690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hypoxia is a common feature of solid tumors and is associated with increased tumor progression, resistance to therapy and increased metastasis. Hence, tumor hypoxia is a prognostic factor independent of treatment modality. To survive hypoxia, cells activate macroautophagy/autophagy. Paradoxically, in several cancer types, mutations or loss of essential autophagy genes have been reported that are associated with earlier onset of tumor growth. However, to our knowledge, the phenotypic and therapeutic consequences of autophagy deficiency have remained unexplored. In this study, we determined autophagy-defects in head and neck squamous cell carcinoma (HNSCC) and observed that expression of ATG12 (autophagy related 12) was lost in 25%-40% of HNSCC. In line, ATG12 loss is associated with absence of hypoxia, as determined by pimonidazole immunohistochemistry. Hence, ATG12 loss is associated with improved prognosis after therapy in two independent HNSCC cohorts and 7 additional cancer types. In vivo, ATG12 targeting resulted in decreased hypoxia tolerance, increased necrosis and sensitivity of the tumor to therapy, but in vitro ATG12-deficient cells displayed enhanced survival in nutrient-rich culture medium. Besides oxygen, delivery of glucose was hampered in hypoxic regions in vivo, which increases the reliance of cells on other carbon sources (e.g., L-glutamine). We observed decreased intracellular L-glutamine levels in ATG12-deficient cells during hypoxia and increased cell killing after L-glutamine depletion, indicating a central role for ATG12 in maintaining L-glutamine homeostasis. Our results demonstrate that ATG12low tumors represent a phenotypically different subtype that, due to the lowered hypoxia tolerance, display a favorable outcome after therapy. Abbreviations: ARCON:accelerated radiotherapy with carbogen and nicotinamide; ATG: autophagy related; BrdUrd: bromodeoxyuridine; CA9/CAIX: carbonic anhydrase 9; HIF1A/HIF1α: hypoxia inducible factor 1 subunit alpha; HNSCC: head and neck squamous cell carcinoma; HPV: human papilloma virus; HR: hazard ratio; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MEF: mouse embryonic fibroblast; mRNA: messenger ribonucleic acid; PCR: polymerase chain reaction; SLC2A1/GLUT1: solute carrier family 2 member 1; TCGA: the Cancer Genome Atlas; TME: tumor microenvironment; UTR: untranslated region; VEGF: vascular endothelial growth factor
Collapse
Affiliation(s)
- Tom G Keulers
- Department of Radiotherapy, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Alexander Koch
- Department of Pathology, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marike W van Gisbergen
- The M-Lab, Department of Precision Medicine, Grow - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Lydie M O Barbeau
- Department of Radiotherapy, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marijke I Zonneveld
- Department of Radiotherapy, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Monique C de Jong
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kim G M Savelkouls
- Department of Radiotherapy, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Veerle Melotte
- Department of Pathology, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Kasper M A Rouschop
- Department of Radiotherapy, Grow - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
72
|
Biondo M, Panuzzo C, Ali SM, Bozzaro S, Osella M, Bracco E, Pergolizzi B. The Dynamics of Aerotaxis in a Simple Eukaryotic Model. Front Cell Dev Biol 2021; 9:720623. [PMID: 34888305 PMCID: PMC8650612 DOI: 10.3389/fcell.2021.720623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/13/2021] [Indexed: 01/09/2023] Open
Abstract
In aerobic organisms, oxygen is essential for efficient energy production, and it acts as the last acceptor of the mitochondrial electron transport chain and as regulator of gene expression. However, excessive oxygen can lead to production of deleterious reactive oxygen species. Therefore, the directed migration of single cells or cell clumps from hypoxic areas toward a region of optimal oxygen concentration, named aerotaxis, can be considered an adaptive mechanism that plays a major role in biological and pathological processes. One relevant example is the development of O2 gradients when tumors grow beyond their vascular supply, leading frequently to metastasis. In higher eukaryotic organisms, aerotaxis has only recently begun to be explored, but genetically amenable model organisms suitable to dissect this process remain an unmet need. In this regard, we sought to assess whether Dictyostelium cells, which are an established model for chemotaxis and other motility processes, could sense oxygen gradients and move directionally in their response. By assessing different physical parameters, our findings indicate that both growing and starving Dictyostelium cells under hypoxic conditions migrate directionally toward regions of higher O2 concentration. This migration is characterized by a specific pattern of cell arrangement. A thickened circular front of high cell density (corona) forms in the cell cluster and persistently moves following the oxygen gradient. Cells in the colony center, where hypoxia is more severe, are less motile and display a rounded shape. Aggregation-competent cells forming streams by chemotaxis, when confined under hypoxic conditions, undergo stream or aggregate fragmentation, giving rise to multiple small loose aggregates that coordinately move toward regions of higher O2 concentration. By testing a panel of mutants defective in chemotactic signaling, and a catalase-deficient strain, we found that the latter and the pkbR1null exhibited altered migration patterns. Our results suggest that in Dictyostelium, like in mammalian cells, an intracellular accumulation of hydrogen peroxide favors the migration toward optimal oxygen concentration. Furthermore, differently from chemotaxis, this oxygen-driven migration is a G protein-independent process.
Collapse
Affiliation(s)
- Marta Biondo
- Department of Physics, INFN, University of Turin, Turin, Italy
| | - Cristina Panuzzo
- Department of Clinical and Biological Science, University of Turin, Turin, Italy
| | - Shahzad M Ali
- Department of Clinical and Biological Science, University of Turin, Turin, Italy
| | - Salvatore Bozzaro
- Department of Clinical and Biological Science, University of Turin, Turin, Italy
| | - Matteo Osella
- Department of Physics, INFN, University of Turin, Turin, Italy
| | - Enrico Bracco
- Department of Oncology, University of Turin, Turin, Italy
| | - Barbara Pergolizzi
- Department of Clinical and Biological Science, University of Turin, Turin, Italy
| |
Collapse
|
73
|
Maitz CA, Tate D, Bechtel S, Lunceford J, Henry C, Flesner B, Collins A, Varterasian M, Tung D, Zhang L, Saha S, Bryan JN. Paired 18F-Fluorodeoxyglucose (18F-FDG), and 64Cu-Copper(II)-diacetyl-bis(N(4)-methylthiosemicarbazone) (64Cu-ATSM) PET Scans in Dogs with Spontaneous Tumors and Evaluation for Hypoxia-Directed Therapy. Radiat Res 2021; 197:253-260. [PMID: 34855934 DOI: 10.1667/rade-20-00186.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/01/2021] [Indexed: 11/03/2022]
Abstract
Hypoxia is associated with neoplastic tissue, protecting cancer cells from death by irradiation and chemotherapy. Identification of hypoxic volume of tumors could optimize patient selection for hypoxia-directed medical, immunological, and radiation therapies. Clostridium novyi-NT (CNV-NT) is an oncolytic bacterium derived from attenuated wild-type Clostridium novyi spores, which germinates exclusively in the anaerobic core of tumors with low-oxygen content. The hypothesis was that 64Cu-ATSM would localize to regions of hypoxia, and that greater hypoxic volume would result in greater germination of Clostridium novyi-NT (CNV-NT). Tumor-bearing companion dogs were recruited to a veterinary clinical trial. Dogs received a CT scan, 18F-FDG PET scan (74 MBq) and 64Cu-ATSM PET scan (74 MBq). Scan regions of interest were defined as the highest 20% of counts/voxel for each PET scan, and regions with voxels overlapping between the two scans. Maximum standardized uptake value (MaxSUV) and threshold volume were calculated. Direct oximetry was performed in select tumors. Tumor types evaluated included nerve sheath tumor (10), apocrine carcinoma (1), melanoma (3) and oral sarcoma (6). MaxSUVATSM ranged from 0.3-6.6. Measured oxygen tension ranged from 0.05-89.9 mmHg. Inverse of MaxSUVATSM had a linear relationship with oxygen tension (R2 = 0.53, P = 0.0048). Hypoxia <8 mmHg was associated with an SUVATSM > 1.0. Hypoxic volume ranged from 0 to 100% of gross tumor volume (GTV) and MaxSUVATSM was positively correlated with hypoxic volume (R = 0.674; P = 0.0001), but not GTV (P = 0.182). Tumor hypoxic volume was heterogeneous in location and distribution. 64Cu-ATSM-avid regions were associated with differential CT attenuation. Hypoxic volume did not predict CNV-NT germination. 64Cu-ATSM PET scanning predicts hypoxia patterns within spontaneously occurring tumors of dogs as measured by direct oxymetry. Total tumor volume does not accurately predict degree or proportion of tumor hypoxia.
Collapse
Affiliation(s)
- Charles A Maitz
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| | - Deborah Tate
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| | - Sandra Bechtel
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| | - Joni Lunceford
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| | - Carolyn Henry
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| | - Brian Flesner
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| | | | | | - David Tung
- Biomed Valley Discoveries, Inc., Kansas City, Missouri
| | - Linping Zhang
- Biomed Valley Discoveries, Inc., Kansas City, Missouri
| | - Saurabh Saha
- Biomed Valley Discoveries, Inc., Kansas City, Missouri
| | - Jeffrey N Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri - Columbia, Missouri
| |
Collapse
|
74
|
Raufi AG, Liguori NR, Carlsen L, Parker C, Hernandez Borrero L, Zhang S, Tian X, Louie A, Zhou L, Seyhan AA, El-Deiry WS. Therapeutic Targeting of Autophagy in Pancreatic Ductal Adenocarcinoma. Front Pharmacol 2021; 12:751568. [PMID: 34916936 PMCID: PMC8670090 DOI: 10.3389/fphar.2021.751568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease characterized by early metastasis, late detection, and poor prognosis. Progress towards effective therapy has been slow despite significant efforts. Novel treatment approaches are desperately needed and autophagy, an evolutionary conserved process through which proteins and organelles are recycled for use as alternative energy sources, may represent one such target. Although incompletely understood, there is growing evidence suggesting that autophagy may play a role in PDAC carcinogenesis, metastasis, and survival. Early clinical trials involving autophagy inhibiting agents, either alone or in combination with chemotherapy, have been disappointing. Recently, evidence has demonstrated synergy between the MAPK pathway and autophagy inhibitors in PDAC, suggesting a promising therapeutic intervention. In addition, novel agents, such as ONC212, have preclinical activity in pancreatic cancer, in part through autophagy inhibition. We discuss autophagy in PDAC tumorigenesis, metabolism, modulation of the immune response, and preclinical and clinical data with selected autophagy modulators as therapeutics.
Collapse
Affiliation(s)
- Alexander G. Raufi
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- *Correspondence: Wafik S. El-Deiry, ; Alexander G. Raufi,
| | - Nicholas R. Liguori
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Temple University, Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Cassandra Parker
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Surgery, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Liz Hernandez Borrero
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Xiaobing Tian
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Anna Louie
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Surgery, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- *Correspondence: Wafik S. El-Deiry, ; Alexander G. Raufi,
| |
Collapse
|
75
|
Roussakis Y, Anagnostopoulos G. Physical and Dosimetric Aspects of the Iridium-Knife. Front Oncol 2021; 11:728452. [PMID: 34858815 PMCID: PMC8630660 DOI: 10.3389/fonc.2021.728452] [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: 06/21/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
The three-dimensional iridium-192 (192Ir) high-dose-rate (HDR) brachytherapy manifests itself as a high-precision, hypofractionated, dose-escalating, minimally invasive method in the armamentarium of contemporary radiation oncology clinical applications. In this study, the physical aspects of the 192Ir radionuclide are presented. Its dosimetric application in HDR brachytherapy for different anatomical sites (prostate, gynecological malignancies, liver, and intrathoracic tumors) as well as the corresponding dosimetric comparison with the stereotactic body radiation therapy (SBRT) techniques based on a representative selection of dosimetric publications is reviewed and illustrated.
Collapse
|
76
|
Datta A, West C, O'Connor JPB, Choudhury A, Hoskin P. Impact of hypoxia on cervical cancer outcomes. Int J Gynecol Cancer 2021; 31:1459-1470. [PMID: 34593564 DOI: 10.1136/ijgc-2021-002806] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/14/2021] [Indexed: 01/22/2023] Open
Abstract
The annual global incidence of cervical cancer is approximately 604 000 cases/342 000 deaths, making it the fourth most common cancer in women. Cervical cancer is a major healthcare problem in low and middle income countries where 85% of new cases and deaths occur. Secondary prevention measures have reduced incidence and mortality in developed countries over the past 30 years, but cervical cancer remains a major cause of cancer deaths in women. For women who present with Fédération Internationale de Gynécologie et d'Obstétrique (FIGO 2018) stages IB3 or upwards, chemoradiation is the established treatment. Despite high rates of local control, overall survival is less than 50%, largely due to distant relapse. Reducing the health burden of cervical cancer requires greater individualization of treatment, identifying those at risk of relapse and progression for modified or intensified treatment. Hypoxia is a well known feature of solid tumors and an established therapeutic target. Low tumorous oxygenation increases the risk of local invasion, metastasis and treatment failure. While meta-analyses show benefit, many individual trials targeting hypoxia failed in part due to not selecting patients most likely to benefit. This review summarizes the available hypoxia-targeted strategies and identifies further research and new treatment paradigms needed to improve patient outcomes. The applications and limitations of hypoxia biomarkers for treatment selection and response monitoring are discussed. Finally, areas of greatest unmet clinical need are identified to measure and target hypoxia and therefore improve cervical cancer outcomes.
Collapse
Affiliation(s)
- Anubhav Datta
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Radiology, The Christie NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, The Christie Hospital NHS Trust, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| |
Collapse
|
77
|
Vilaplana-Lopera N, Besh M, Moon EJ. Targeting Hypoxia: Revival of Old Remedies. Biomolecules 2021; 11:1604. [PMID: 34827602 PMCID: PMC8615589 DOI: 10.3390/biom11111604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022] Open
Abstract
Tumour hypoxia is significantly correlated with patient survival and treatment outcomes. At the molecular level, hypoxia is a major driving factor for tumour progression and aggressiveness. Despite the accumulative scientific and clinical efforts to target hypoxia, there is still a need to find specific treatments for tumour hypoxia. In this review, we discuss a variety of approaches to alter the low oxygen tumour microenvironment or hypoxia pathways including carbogen breathing, hyperthermia, hypoxia-activated prodrugs, tumour metabolism and hypoxia-inducible factor (HIF) inhibitors. The recent advances in technology and biological understanding reveal the importance of revisiting old therapeutic regimens and repurposing their uses clinically.
Collapse
Affiliation(s)
| | | | - Eui Jung Moon
- Department of Oncology, MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington OX3 7DQ, UK; (N.V.-L.); (M.B.)
| |
Collapse
|
78
|
Kunder S, Chatterjee A, Manna S, Mahimkar M, Patil A, Rangarajan V, Budrukkar A, Ghosh-Laskar S, Agarwal JP, Gupta T. Correlation between imaging and tissue biomarkers of hypoxia in squamous cell cancer of the head and neck. World J Nucl Med 2021; 20:228-236. [PMID: 34703390 PMCID: PMC8488888 DOI: 10.4103/wjnm.wjnm_91_20] [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: 07/04/2020] [Revised: 11/29/2020] [Accepted: 01/11/2021] [Indexed: 11/04/2022] Open
Abstract
The aim of this study was to correlate endogenous tissue biomarkers of hypoxia with quantitative imaging parameters derived from 18F-fluoro-misonidazole (F-MISO) and 18F-fluoro-deoxy-glucose (FDG) positron emission tomography/computed tomography (PET/CT) and clinical outcomes in locoregionally advanced head and neck squamous cell carcinoma (HNSCC). Tumor-tissue blocks of HNSCC patients with pretreatment F-MISO-PET/CT and FDG-PET/CT were de-archived for expression of hypoxia-inducible factor-1 alpha (HIF-1α) subunit, carbonic anhydrase-IX (CA-IX), and glucose transporter subunit-1 (GLUT-1) using immunohistochemistry (IHC). The intensity of staining was graded and correlated with quantitative imaging parameters and with disease-related outcomes. Tissue blocks were analyzed for 14 of 20 patients. On IHC, median H-scores for HIF-1α, CA-IX, and GLUT-1 were 130, 0, and 95, respectively. No significant correlation of tissue biomarkers of hypoxia with quantitative imaging parameters was found. However, borderline significant correlation was seen for H-scores of CA-IX with hypoxic tumor volume (HTV) (r = 0.873, P = 0.054) and fractional hypoxic volume (r = 0.824, P = 0.086) derived from F-MISO-PET/CT. At a median follow-up of 43 months, 5-year Kaplan-Meier estimates of locoregional control, disease-free survival, and overall survival were 53%, 43%, and 40%, respectively. Increased expression of HIF-1α or GLUT-1 (dichotomized by median H-scores) was not individually associated with disease-related outcomes. However, a combination of high HTV (>4.89cc) with above median H-scores of either HIF-1α (>130) and/or GLUT-1 (>95) was associated with worse clinical outcomes. None of the three patients with such "adverse hypoxic profile" were long-term survivors. There is no significant correlation of endogenous tissue biomarkers of hypoxia (HIF-1α, CA-IX, and GLUT-1) with quantitative imaging parameters (on F-MISO-PET/CT and FDG-PET/CT) or long-term outcomes in HNSCC. However, a combination of both can identify a subgroup of patients with adverse outcomes.
Collapse
Affiliation(s)
- Shreya Kunder
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Subhakankha Manna
- Department of Mahimkar Lab, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Manoj Mahimkar
- Department of Mahimkar Lab, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Asawari Patil
- Department of Pathology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine & Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Ashwini Budrukkar
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sarbani Ghosh-Laskar
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Jai Prakash Agarwal
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC)/Tata Memorial Hospital (TMH), Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
79
|
Zhao S, Zhou L, Dicker DT, Lev A, Zhang S, Ross E, El-Deiry WS. Anti-cancer efficacy including Rb-deficient tumors and VHL-independent HIF1α proteasomal destabilization by dual targeting of CDK1 or CDK4/6 and HSP90. Sci Rep 2021; 11:20871. [PMID: 34686682 PMCID: PMC8536770 DOI: 10.1038/s41598-021-00150-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
A prevalent characteristic of solid tumors is intra-tumoral hypoxia. Hypoxia-inducible factor 1α (HIF1α) predominantly mediates the adaptive response to O2 oscillation and is linked to multiple malignant hallmarks. Here we describe a strategy to robustly target HIF1α by dual inhibition of CDK(s) and heat shock protein 90 (HSP90). We show that CDK1 may contribute to HSP90-mediated HIF1α stabilization. CDK1 knockdown enhances the decrease of HIF1α by HSP90 inhibition. Dual inhibition of CDK1 and HSP90 significantly increases apoptosis and synergistically inhibits cancer cell viability. Similarly, targeting CDK4/6 using FDA-approved inhibitors in combination with HSP90 inhibition shows a class effect on HIF1α inhibition and cancer cell viability suppression not only in colorectal but also in various other cancer types, including Rb-deficient cancer cells. Dual inhibition of CDK4/6 and HSP90 suppresses tumor growth in vivo. In summary, combined targeting of CDK(s) (CDK1 or CDK4/6) and HSP90 remarkably inhibits the expression level of HIF1α and shows promising anti-cancer efficacy with therapeutic potential.
Collapse
Affiliation(s)
- Shuai Zhao
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Pathobiology Graduate Program, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - David T Dicker
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA
| | - Avital Lev
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Eric Ross
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA. .,Pathobiology Graduate Program, Brown University, Providence, RI, USA. .,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA. .,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA. .,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA. .,Fox Chase Cancer Center, Philadelphia, PA, USA. .,Hematology/Oncology Division, Lifespan Cancer Institute, Providence, RI, USA.
| |
Collapse
|
80
|
Reffai A, Mesmoudi M, Derkaoui T, Ghailani Nourouti N, Barakat A, Sellal N, Mallick P, Bennani Mechita M. Epidemiological Profile and Clinicopathological, Therapeutic, and Prognostic Characteristics of Nasopharyngeal Carcinoma in Northern Morocco. Cancer Control 2021; 28:10732748211050587. [PMID: 34664512 PMCID: PMC8529313 DOI: 10.1177/10732748211050587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Nasopharyngeal carcinoma is a multifactorial disease mainly affecting the
Asian and North African populations including Morocco. This study aimed to
determine the epidemiological profile of nasopharyngeal carcinoma in
Northern Morocco as well as its clinicopathological, therapeutic, and
prognostic characteristics. Methods 129 patients with nasopharyngeal carcinoma followed at the regional center of
oncology of Tangier in the period between April 2017 and July 2019, and
diagnosed elsewhere from March 2000 to February 2019, were included in this
study. Statistical analysis of the data was realized using Statistical
Package for the Social Sciences (SPSS) software. Results Nasopharyngeal carcinoma (NPC) represented 5% of all cases with a median age
of 50. The most affected age group was 40–54 years (41.1%). Of all patients,
65.9% were men and 34.1% were women with a sex ratio of 1.93 (Male/Female).
Undifferentiated nasopharyngeal carcinomas were the most common histological
type affecting 96.12% of patients. At diagnosis, the majority of patients
(82.2%) had an advanced stage of NPC (III, VIa, b, c) including 5.4% of
metastatic cases (IVc). Most cases (86%) had lymph node involvement with
cervical mass being the most common clinical presentation. 81.4% of patients
received radiotherapy combined with chemotherapy. Among these patients,
54.3% had concurrent radiochemotherapy preceded by induction chemotherapy.
The 5-year overall survival (OS) was 86.8% for all patients. It represented
91.3% for early stages, 87.9% for locally advanced stages, and 57.1% for the
metastatic stage significantly. The disease-free survival (DFS) at 5 years
was 87.6% knowing that relapse occurred in 16 cases. Conclusions Nasopharyngeal carcinoma is a particular disease with a late declaration. It
is common in Morocco as is the case in other endemic areas with a high
prevalence. Patients’ survival is significantly influenced by disease
staging.
Collapse
Affiliation(s)
- Ayman Reffai
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Science and Technology of Tangier (FSTT), 531748Abdelmalek Essaadi University (UAE), Tangier, Morocco
| | - Mohamed Mesmoudi
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Science and Technology of Tangier (FSTT), 531748Abdelmalek Essaadi University (UAE), Tangier, Morocco.,Ahmed Ben Zayed Al Nahyan Center of Cancer Treatment, Tangier, Morocco
| | - Touria Derkaoui
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Science and Technology of Tangier (FSTT), 531748Abdelmalek Essaadi University (UAE), Tangier, Morocco
| | - Naima Ghailani Nourouti
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Science and Technology of Tangier (FSTT), 531748Abdelmalek Essaadi University (UAE), Tangier, Morocco
| | - Amina Barakat
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Science and Technology of Tangier (FSTT), 531748Abdelmalek Essaadi University (UAE), Tangier, Morocco
| | - Nabila Sellal
- Ahmed Ben Zayed Al Nahyan Center of Cancer Treatment, Tangier, Morocco
| | - Parag Mallick
- Canary Center for Cancer Early Detection, School of Medicine, 10624Stanford University, Stanford, CA, USA
| | - Mohcine Bennani Mechita
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Science and Technology of Tangier (FSTT), 531748Abdelmalek Essaadi University (UAE), Tangier, Morocco
| |
Collapse
|
81
|
Chen EY, Tse D, Hou H, Schreiber WA, Schaner PE, Kmiec MM, Hebert KA, Kuppusamy P, Swartz HM, Williams BB. Evaluation of a Refined Implantable Resonator for Deep-Tissue EPR Oximetry in the Clinic. APPLIED MAGNETIC RESONANCE 2021; 52:1321-1342. [PMID: 34744319 PMCID: PMC8570533 DOI: 10.1007/s00723-021-01376-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 05/04/2023]
Abstract
OBJECTIVES (1) Summarize revisions made to the implantable resonator (IR) design and results of testing to characterize biocompatibility;(2) Demonstrate safety of implantation and feasibility of deep tissue oxygenation measurement using electron paramagnetic resonance (EPR) oximetry. STUDY DESIGN In vitro testing of the revised IR and in vivo implantation in rabbit brain and leg tissues. METHODS Revised IRs were fabricated with 1-4 OxyChips with a thin wire encapsulated with two biocompatible coatings. Biocompatibility and chemical characterization tests were performed. Rabbits were implanted with either an IR with 2 oxygen sensors or a biocompatible-control sample in both the brain and hind leg. The rabbits were implanted with IRs using a catheter-based, minimally invasive surgical procedure. EPR oximetry was performed for rabbits with IRs. Cohorts of rabbits were euthanized and tissues were obtained at 1 week, 3 months, and 9 months after implantation and examined for tissue reaction. RESULTS Biocompatibility and toxicity testing of the revised IRs demonstrated no abnormal reactions. EPR oximetry from brain and leg tissues were successfully executed. Blood work and histopathological evaluations showed no significant difference between the IR and control groups. CONCLUSIONS IRs were functional for up to 9 months after implantation and provided deep tissue oxygen measurements using EPR oximetry. Tissues surrounding the IRs showed no more tissue reaction than tissues surrounding the control samples. This pre-clinical study demonstrates that the IRs can be safely implanted in brain and leg tissues and that repeated, non-invasive, deep-tissue oxygen measurements can be obtained using in vivo EPR oximetry.
Collapse
Affiliation(s)
- Eunice Y. Chen
- Section of Otolaryngology, Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States and Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Dan Tse
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Huagang Hou
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Wilson A. Schreiber
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Philip E. Schaner
- Section of Radiation Oncology, Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States and Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Maciej M. Kmiec
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Kendra A. Hebert
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Periannan Kuppusamy
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Harold M. Swartz
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Section of Radiation Oncology, Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States and Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Benjamin B. Williams
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Section of Radiation Oncology, Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States and Geisel School of Medicine at Dartmouth, Hanover, NH
| |
Collapse
|
82
|
Huang Y, Fan J, Li Y, Fu S, Chen Y, Wu J. Imaging of Tumor Hypoxia With Radionuclide-Labeled Tracers for PET. Front Oncol 2021; 11:731503. [PMID: 34557414 PMCID: PMC8454408 DOI: 10.3389/fonc.2021.731503] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/19/2021] [Indexed: 01/27/2023] Open
Abstract
The hypoxic state in a solid tumor refers to the internal hypoxic environment that appears as the tumor volume increases (the maximum radius exceeds 180-200 microns). This state can promote angiogenesis, destroy the balance of the cell’s internal environment, and lead to resistance to radiotherapy and chemotherapy, as well as poor prognostic factors such as metastasis and recurrence. Therefore, accurate quantification, mapping, and monitoring of hypoxia, targeted therapy, and improvement of tumor hypoxia are of great significance for tumor treatment and improving patient survival. Despite many years of development, PET-based hypoxia imaging is still the most widely used evaluation method. This article provides a comprehensive overview of tumor hypoxia imaging using radionuclide-labeled PET tracers. We introduced the mechanism of tumor hypoxia and the reasons leading to the poor prognosis, and more comprehensively included the past, recent and ongoing studies of PET radiotracers for tumor hypoxia imaging. At the same time, the advantages and disadvantages of mainstream methods for detecting tumor hypoxia are summarized.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Junying Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, China
| | - Yue Chen
- Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, China.,Nuclear Medicine and Molecular Imaging key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, China
| |
Collapse
|
83
|
Liu J, Chen J, Liu H, Zhang K, Zeng Q, Yang S, Jiang Z, Zhang X, Chen T, Li D, Shan H. Bi/Se-Based Nanotherapeutics Sensitize CT Image-Guided Stereotactic Body Radiotherapy through Reprogramming the Microenvironment of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42473-42485. [PMID: 34474563 DOI: 10.1021/acsami.1c11763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The particular characteristics of hypoxia, immune suppression in the tumor microenvironment, and the lack of accurate imaging guidance lead to the limited effects of stereotactic body radiotherapy (SBRT) in reducing the recurrence rate and mortality of hepatocellular carcinoma (HCC). This research developed a novel theranostic agent based on Bi/Se nanoparticles (NPs), synthesized by a simple reduction reaction method for in vivo CT image-guided SBRT sensitization in mice. After loading Lenvatinib (Len), the obtained Bi/Se-Len NPs had excellent performance in reversing hypoxia and the immune suppression status of HCC. In vivo CT imaging results uncovered that the radiotherapy (RT) area could be accurately labeled after the injection of Bi/Se-Len NPs. Under Len's unique and robust properties, in vivo treatment was then carried out upon injection of Bi/Se-Len NPs, achieving excellent RT sensitization effects in a mouse HCC model. Comprehensive tests and histological stains revealed that Bi/Se-Len NPs could reshape and normalize tumor blood vessels, reduce the hypoxic situation of the tumor, and upregulate tumor-infiltrating CD4+ and CD8+ T lymphocytes around the tumors. Our work highlights an excellent proposal of Bi/Se-Len NPs as theranostic nanoparticles for image-guided HCC radiotherapy.
Collapse
Affiliation(s)
- Jiani Liu
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- The Cancer Center of The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Jiayao Chen
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Hongxing Liu
- Department of Chemistry, Jinan University, Guangzhou, Guangdong Province, 510632, P. R. China
| | - Ke Zhang
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Qi Zeng
- The Cancer Center of The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Shuai Yang
- The Cancer Center of The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Xiaoting Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, Guangdong Province, 510632, P. R. China
| | - Dan Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Hong Shan
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| |
Collapse
|
84
|
Colombani T, Eggermont LJ, Hatfield SM, Rogers ZJ, Rezaeeyazdi M, Memic A, Sitkovsky MV, Bencherif SA. Oxygen-Generating Cryogels Restore T Cell Mediated Cytotoxicity in Hypoxic Tumors. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2102234. [PMID: 37745940 PMCID: PMC10516343 DOI: 10.1002/adfm.202102234] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 09/26/2023]
Abstract
Solid tumors are protected from antitumor immune responses due to their hypoxic microenvironments. Weakening hypoxia-driven immunosuppression by hyperoxic breathing of 60% oxygen has shown to be effective in unleashing antitumor immune cells against solid tumors. However, efficacy of systemic oxygenation is limited against solid tumors outside of lungs and has been associated with unwanted side effects. As a result, it is essential to develop targeted oxygenation alternatives to weaken tumor hypoxia as novel approaches to restore immune responses against cancer. Herein, we report on injectable oxygen-generating cryogels (O2-cryogels) to reverse tumor-induced hypoxia. These macroporous biomaterials were designed to locally deliver oxygen, inhibit the expression of hypoxia-inducible genes in hypoxic melanoma cells, and reduce the accumulation of immunosuppressive extracellular adenosine. Our data show that O2-cryogels enhance T cell-mediated secretion of cytotoxic proteins, restoring the killing ability of tumor-specific CTLs, both in vitro and in vivo. In summary, O2-cryogels provide a unique and safe platform to supply oxygen as a co-adjuvant in hypoxic tumors and have the potential to improve cancer immunotherapies.
Collapse
Affiliation(s)
- Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Loek J. Eggermont
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Stephen M. Hatfield
- New England Inflammation and Tissue Protection Institute, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Zachary J. Rogers
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | | | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah, Makkah 21589, Saudi Arabia
| | - Michail V. Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Biomechanics and Bioengineering (BMBI), UTC CNRS UMR 7338, University of Technology of Compiègne, Sorbonne University, 60203 Compiègne, France
| |
Collapse
|
85
|
Stanislavova N, Karamanliev M, Ivanov T, Yotsov T, Zhou K, Dimitrov D. Is high-intensity focused ultrasound (HIFU) an option for neoadjuvant therapy for borderline resectable pancreatic cancer patients? - a systematic review. Int J Hyperthermia 2021; 38:75-80. [PMID: 34420446 DOI: 10.1080/02656736.2021.1909150] [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] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Pancreatic cancer is with the poorest prognosis of all common cancers worldwide. Despite the advances in treatment the results are poor throughout the different methods. Pancreatic resection still yields the best outcome. However only a quarter of the patients present at operable stage. HIFU is a noninvasive technique that can be used to treat pancreatic cancer. AIM The aim of this review is to perform a systematic review on the data about the resection rate after HIFU ablation in patients with borderline resectable pancreatic cancer (BRPC) and the impact of this technique over the oncological results. MATERIALS AND METHODS The PubMed and Wanfang databases were searched using keywords: pancreatic cancer, HIFU ablation and high-intensity focused ultrasound. All found articles were reviewed. The systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standard guidelines. This study was financially supported by 2019 'Kuan-Ren Elite' Program of 2nd Affiliated Hospital of Chongqing Medical University, China (Grant no. KY2019G019). RESULTS The English database search showed 109 papers, of which 3 met the inclusion criteria. The Wanfang database resulted in 110 papers; however, none met the inclusion criteria of the review. From the included studies 97 patients underwent neoadjuvant HIFU ablation ± chemotherapy. Thirty-four patients reached resection (35.1%). In two patients, residual tumor (R) classification was not reported. R0 resection rate in all reported patients is 30.5% (29/95). R1 resection rate is 3.2% (3/95). CONCLUSION HIFU is found to be safe and feasible in locally advanced and metastatic pancreatic cancer with proven downstaging and downsizing effects. Further research on role of HIFU ablation as a neoadjuvant treatment for borderline resectable pancreatic cancer is needed.
Collapse
Affiliation(s)
- Nadya Stanislavova
- Faculty of Medicine, HIFU Center, University St. Marina Hospital, Medical University, Pleven, Bulgaria
| | - Martin Karamanliev
- Department of Surgical Oncology, Faculty of Medicine, Medical University, Pleven, Bulgaria
| | - Tsvetomir Ivanov
- Department of Surgical Oncology, Faculty of Medicine, Medical University, Pleven, Bulgaria
| | - Tsanko Yotsov
- Department of Surgical Oncology, Faculty of Medicine, Medical University, Pleven, Bulgaria
| | - Kun Zhou
- Clinical Center for Tumor Therapy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dobromir Dimitrov
- Faculty of Medicine, HIFU Center, University St. Marina Hospital, Medical University, Pleven, Bulgaria.,Department of Surgical Oncology, Faculty of Medicine, Medical University, Pleven, Bulgaria
| |
Collapse
|
86
|
Sebeke LC, Rademann P, Maul AC, Yeo SY, Castillo Gómez JD, Deenen DA, Schmidt P, de Jager B, Heemels WPMH, Grüll H, Heijman E. Visualization of thermal washout due to spatiotemporally heterogenous perfusion in the application of a model-based control algorithm for MR-HIFU mediated hyperthermia. Int J Hyperthermia 2021; 38:1174-1187. [PMID: 34374624 DOI: 10.1080/02656736.2021.1933616] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE This article will report results from the in-vivo application of a previously published model-predictive control algorithm for MR-HIFU hyperthermia. The purpose of the investigation was to test the controller's in-vivo performance and behavior in the presence of heterogeneous perfusion. MATERIALS AND METHODS Hyperthermia at 42°C was induced and maintained for up to 30 min in a circular section of a thermometry slice in the biceps femoris of German landrace pigs (n=5) using a commercial MR-HIFU system and a recently developed MPC algorithm. The heating power allocation was correlated with heat sink maps and contrast-enhanced MRI images. The temporal change in perfusion was estimated based on the power required to maintain hyperthermia. RESULTS The controller performed well throughout the treatments with an absolute average tracking error of 0.27 ± 0.15 °C and an average difference of 1.25 ± 0.22 °C between T10 and T90. The MPC algorithm allocates additional heating power to sub-volumes with elevated heat sink effects, which are colocalized with blood vessels visible on contrast-enhanced MRI. The perfusion appeared to have increased by at least a factor of ∼1.86 on average. CONCLUSIONS The MPC controller generates temperature distributions with a narrow spectrum of voxel temperatures inside the target ROI despite the presence of spatiotemporally heterogeneous perfusion due to the rapid thermometry feedback available with MR-HIFU and the flexible allocation of heating power. The visualization of spatiotemporally heterogeneous perfusion presents new research opportunities for the investigation of stimulated perfusion in hypoxic tumor regions.
Collapse
Affiliation(s)
- Lukas Christian Sebeke
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, Cologne, Germany.,Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, The Netherlands
| | - Pia Rademann
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Experimental Medicine, Cologne, Germany
| | - Alexandra Claudia Maul
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Experimental Medicine, Cologne, Germany
| | - Sin Yuin Yeo
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, Cologne, Germany.,Profound Medical GmbH, Hamburg, Germany
| | - Juan Daniel Castillo Gómez
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, Cologne, Germany
| | - Daniel A Deenen
- Eindhoven University of Technology, Department of Mechanical Engineering, Control Systems Technology, Eindhoven, The Netherlands
| | - Patrick Schmidt
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, Cologne, Germany
| | - Bram de Jager
- Eindhoven University of Technology, Department of Mechanical Engineering, Control Systems Technology, Eindhoven, The Netherlands
| | - W P M H Heemels
- Eindhoven University of Technology, Department of Mechanical Engineering, Control Systems Technology, Eindhoven, The Netherlands
| | - Holger Grüll
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, Cologne, Germany.,Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, The Netherlands
| | - Edwin Heijman
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, Cologne, Germany.,Philips Research, Eindhoven, The Netherlands
| |
Collapse
|
87
|
Liu J, Cabral H, Song B, Aoki I, Chen Z, Nishiyama N, Huang Y, Kataoka K, Mi P. Nanoprobe-Based Magnetic Resonance Imaging of Hypoxia Predicts Responses to Radiotherapy, Immunotherapy, and Sensitizing Treatments in Pancreatic Tumors. ACS NANO 2021; 15:13526-13538. [PMID: 34355882 DOI: 10.1021/acsnano.1c04263] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Accurate diagnosis of tumors and predicting the therapeutic responses are highly demanded in the clinic to improve the treatment efficacy and survival rates. Since hypoxia develops in the progression of tumors and inversely correlates with prognosis and promotes resistance to radiotherapies and immunotherapies, it is a potential marker for therapeutic prediction. Therefore, effective discrimination of tumor hypoxia for predicting therapeutic outcomes is critical. Here, a magnetic resonance imaging (MRI)-based diagnosis strategy using contrast-amplifying nanoprobes that sense tumor acidosis and real-time observation of hypoxic conditions in tumors has been developed, aiming at accurate detection of pancreatic tumors and prediction of therapeutic effects. Our approach selectively probed xenograft, allograft, and transgenic spontaneous models of intractable pancreatic cancer, which lacks standardized predictive markers to identify patients who benefit most from treatments, and effectively discriminated the intratumoral hypoxia levels. By stratification of pancreatic tumors based on quantitative MR imaging of hypoxia, it enabled prediction of the responses to radiotherapy and immune checkpoint inhibitors. Moreover, the nanoprobe-based MRI could monitor hypoxia reduction by tumor normalization treatments, which permits visualizing pancreatic tumors that will respond to immune checkpoint blockade therapy, enhancing the response rate. The results demonstrate the potential of our strategy for accurate tumor diagnosis, patient stratification, and effective therapy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Bin Song
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Ichio Aoki
- National Institute of Radiological Sciences, Japan Agency for Quantum and Radiological Science and Technology, Anagawa 4-9-1,
Inage, Chiba 263-8555, Japan
| | - Zhouyun Chen
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259 Nagatsuta, Midori-ku, Yokohama 226-8503, Japan
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| |
Collapse
|
88
|
Li Y, Zhao L, Li XF. Targeting Hypoxia: Hypoxia-Activated Prodrugs in Cancer Therapy. Front Oncol 2021; 11:700407. [PMID: 34395270 PMCID: PMC8358929 DOI: 10.3389/fonc.2021.700407] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022] Open
Abstract
Hypoxia is an important characteristic of most solid malignancies, and is closely related to tumor prognosis and therapeutic resistance. Hypoxia is one of the most important factors associated with resistance to conventional radiotherapy and chemotherapy. Therapies targeting tumor hypoxia have attracted considerable attention. Hypoxia-activated prodrugs (HAPs) are bioreductive drugs that are selectively activated under hypoxic conditions and that can accurately target the hypoxic regions of solid tumors. Both single-agent and combined use with other drugs have shown promising antitumor effects. In this review, we discuss the mechanism of action and the current preclinical and clinical progress of several of the most widely used HAPs, summarize their existing problems and shortcomings, and discuss future research prospects.
Collapse
Affiliation(s)
- Yue Li
- Department of Nuclear Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Long Zhao
- Department of Nuclear Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xiao-Feng Li
- Department of Nuclear Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
89
|
Li X, Wu Y, Zhang R, Bai W, Ye T, Wang S. Oxygen-Based Nanocarriers to Modulate Tumor Hypoxia for Ameliorated Anti-Tumor Therapy: Fabrications, Properties, and Future Directions. Front Mol Biosci 2021; 8:683519. [PMID: 34277702 PMCID: PMC8281198 DOI: 10.3389/fmolb.2021.683519] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/10/2021] [Indexed: 12/27/2022] Open
Abstract
Over the past five years, oxygen-based nanocarriers (NCs) to boost anti-tumor therapy attracted tremendous attention from basic research and clinical practice. Indeed, tumor hypoxia, caused by elevated proliferative activity and dysfunctional vasculature, is directly responsible for the less effectiveness or ineffective of many conventional therapeutic modalities. Undeniably, oxygen-generating NCs and oxygen-carrying NCs can increase oxygen concentration in the hypoxic area of tumors and have also been shown to have the ability to decrease the expression of drug efflux pumps (e.g., P-gp); to increase uptake by tumor cells; to facilitate the generation of cytotoxic reactive oxide species (ROS); and to evoke systematic anti-tumor immune responses. However, there are still many challenges and limitations that need to be further improved. In this review, we first discussed the mechanisms of tumor hypoxia and how it severely restricts the therapeutic efficacy of clinical treatments. Then an up-to-date account of recent progress in the fabrications of oxygen-generating NCs and oxygen-carrying NCs are systematically introduced. The improved physicochemical and surface properties of hypoxia alleviating NCs for increasing the targeting ability to hypoxic cells are also elaborated with special attention to the latest nano-technologies. Finally, the future directions of these NCs, especially towards clinical translation, are proposed. Therefore, we expect to provide some valued enlightenments and proposals in engineering more effective oxygen-based NCs in this promising field in this comprehensive overview.
Collapse
Affiliation(s)
- Xianqiang Li
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yue Wu
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Zhang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Wei Bai
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tiantian Ye
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Shujun Wang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
90
|
Hannon G, Tansi FL, Hilger I, Prina‐Mello A. The Effects of Localized Heat on the Hallmarks of Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
91
|
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S, Zhou H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther 2021; 6:218. [PMID: 34108441 PMCID: PMC8190181 DOI: 10.1038/s41392-021-00641-0] [Citation(s) in RCA: 384] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ye Wang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qingxiang Zeng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuzhi Deng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
92
|
Macedo-Silva C, Benedetti R, Ciardiello F, Cappabianca S, Jerónimo C, Altucci L. Epigenetic mechanisms underlying prostate cancer radioresistance. Clin Epigenetics 2021; 13:125. [PMID: 34103085 PMCID: PMC8186094 DOI: 10.1186/s13148-021-01111-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/02/2021] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT) is one of the mainstay treatments for prostate cancer (PCa), a highly prevalent neoplasm among males worldwide. About 30% of newly diagnosed PCa patients receive RT with a curative intent. However, biochemical relapse occurs in 20–40% of advanced PCa treated with RT either alone or in combination with adjuvant-hormonal therapy. Epigenetic alterations, frequently associated with molecular variations in PCa, contribute to the acquisition of a radioresistant phenotype. Increased DNA damage repair and cell cycle deregulation decreases radio-response in PCa patients. Moreover, the interplay between epigenome and cell growth pathways is extensively described in published literature. Importantly, as the clinical pattern of PCa ranges from an indolent tumor to an aggressive disease, discovering specific targetable epigenetic molecules able to overcome and predict PCa radioresistance is urgently needed. Currently, histone-deacetylase and DNA-methyltransferase inhibitors are the most studied classes of chromatin-modifying drugs (so-called ‘epidrugs’) within cancer radiosensitization context. Nonetheless, the lack of reliable validation trials is a foremost drawback. This review summarizes the major epigenetically induced changes in radioresistant-like PCa cells and describes recently reported targeted epigenetic therapies in pre-clinical and clinical settings. ![]()
Collapse
Affiliation(s)
- Catarina Macedo-Silva
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy.,Cancer Biology and Epigenetics Group, Research Center at Portuguese Oncology Institute of Porto, F Bdg, 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center at Portuguese Oncology Institute of Porto, F Bdg, 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Department of Pathology and Molecular Immunology at School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal.
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy.
| |
Collapse
|
93
|
Fernandes SG, Shah P, Khattar E. Recent Advances in Therapeutic Application of DNA Damage Response Inhibitors against Cancer. Anticancer Agents Med Chem 2021; 22:469-484. [PMID: 34102988 DOI: 10.2174/1871520621666210608105735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
DNA integrity is continuously challenged by intrinsic cellular processes and environmental agents. To overcome this genomic damage, cells have developed multiple signaling pathways collectively named as DNA damage response (DDR) and composed of three components: (i) sensor proteins, which detect DNA damage, (ii) mediators that relay the signal downstream and recruit the repair machinery, and (iii) the repair proteins, which restore the damaged DNA. A flawed DDR and failure to repair the damage lead to the accumulation of genetic lesions and increased genomic instability, which is recognized as a hallmark of cancer. Cancer cells tend to harbor increased mutations in DDR genes and often have fewer DDR pathways than normal cells. This makes cancer cells more dependent on particular DDR pathways and thus become more susceptible to compounds inhibiting those pathways compared to normal cells, which have all the DDR pathways intact. Understanding the roles of different DDR proteins in the DNA damage response and repair pathways and identification of their structures have paved the way for the development of their inhibitors as targeted cancer therapy. In this review, we describe the major participants of various DDR pathways, their significance in carcinogenesis, and focus on the inhibitors developed against several key DDR proteins.
Collapse
Affiliation(s)
- Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| | - Prachi Shah
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| |
Collapse
|
94
|
Shimomura O, Oda T, Hashimoto S, Doi M, Hiroshima Y, Numajiri H, Takahashi K, Furuya K, Miyazaki Y, Owada Y, Ogawa K, Ohara Y, Hisakura K, Akashi Y, Enomoto T, Sakurai H. Survival impact on triple-modal strategy comprising hyperthermia, external radiation, and chemotherapy for unresectable locally advanced (UR-LA) pancreatic ductal adenocarcinoma. Surg Oncol 2021; 37:101542. [PMID: 33740629 DOI: 10.1016/j.suronc.2021.101542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/24/2021] [Accepted: 03/02/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Present treatment strategy for unresectable locally advanced (UR-LA) pancreatic ductal adenocarcinoma (PDAC) patients is controversial. Hence, a triple-modal therapy, which is a multidisciplinary strategy, was designed for patients with UR-LA PDAC by adding hyperthermia to conventional chemoradiotherapy at our institution. In this study we aimed to evaluate the effectiveness of this strategy. METHODS Data of 21 UR-LA PDAC patients who underwent the triple-modal treatment were retrospectively analyzed for evaluating the safety and oncological effect of the treatment. The treatment schedule included, five concurrent infusions of gemcitabine (800 mg/m2) followed by hyperthermia (1 h) and X-ray (2 Gy) or proton beam radiation (2.7 Gy) on days 1, 8, 15, 29, and 36. Additional radiotherapies applied a total dose of 50 Gy/25 fr for X-ray radiation or 67.5 Gy/25 fr for proton beam radiation. RESULTS Median overall survival (OS) was 23.6 months. Conversion surgery was performed in 5 patients (23.8%), and a R0 margin could be achieved in 4 of them; however, their median OS (16.3 months) tended to be shorter than that of the patients who did not undergo resection (23.6 months, p = 0.562). Further, the median OS of patients who underwent proton beam radiation (28.0 months) was significantly longer than that of patients who underwent X-ray radiation (13.9 months, p = 0.045). Most adverse events were manageable, except for one grade 3 gastric ulcer. The median tumor size and marker reduction rates were -17% and -91%, respectively. The tumor responses were partial response, stable disease, and progressive disease in 3, 15, and 3 patients, respectively. CONCLUSION Triple-modal strategy, especially when combined with proton beam radiation, is feasible and results in favorable survival outcomes in patients with UR-LA PDAC.
Collapse
Affiliation(s)
- Osamu Shimomura
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| | - Shinji Hashimoto
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Manami Doi
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuichi Hiroshima
- Department of Radiation Oncology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Haruko Numajiri
- Department of Radiation Oncology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kazuhiro Takahashi
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kinji Furuya
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshihiro Miyazaki
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yohei Owada
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Koichi Ogawa
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yusuke Ohara
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Katsuji Hisakura
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshimasa Akashi
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Enomoto
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hideyuki Sakurai
- Department of Radiation Oncology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
95
|
Corroyer-Dulmont A, Valable S, Fantin J, Chatre L, Toutain J, Teulier S, Bazille C, Letissier E, Levallet J, Divoux D, Ibazizène M, Guillouet S, Perrio C, Barré L, Serres S, Sibson NR, Chapon F, Levallet G, Bernaudin M. Multimodal evaluation of hypoxia in brain metastases of lung cancer and interest of hypoxia image-guided radiotherapy. Sci Rep 2021; 11:11239. [PMID: 34045576 PMCID: PMC8159969 DOI: 10.1038/s41598-021-90662-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023] Open
Abstract
Lung cancer patients frequently develop brain metastases (BM). Despite aggressive treatment including neurosurgery and external-radiotherapy, overall survival remains poor. There is a pressing need to further characterize factors in the microenvironment of BM that may confer resistance to radiotherapy (RT), such as hypoxia. Here, hypoxia was first evaluated in 28 biopsies from patients with non‑small cell lung cancer (NSCLC) BM, using CA-IX immunostaining. Hypoxia characterization (pimonidazole, CA-IX and HIF-1α) was also performed in different preclinical NSCLC BM models induced either by intracerebral injection of tumor cells (H2030-Br3M, H1915) into the cortex and striatum, or intracardial injection of tumor cells (H2030-Br3M). Additionally, [18F]-FMISO-PET and oxygen-saturation-mapping-MRI (SatO2-MRI) were carried out in the intracerebral BM models to further characterize tumor hypoxia and evaluate the potential of Hypoxia-image-guided-RT (HIGRT). The effect of RT on proliferation of BM ([18F]-FLT-PET), tumor volume and overall survival was determined. We showed that hypoxia is a major yet heterogeneous feature of BM from lung cancer both preclinically and clinically. HIGRT, based on hypoxia heterogeneity observed between cortical and striatal metastases in the intracerebrally induced models, showed significant potential for tumor control and animal survival. These results collectively highlight hypoxia as a hallmark of BM from lung cancer and the value of HIGRT in better controlling tumor growth.
Collapse
Affiliation(s)
- Aurélien Corroyer-Dulmont
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
- Medical Physics Department, CLCC François Baclesse, 14000, Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Jade Fantin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Laurent Chatre
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Jérôme Toutain
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Sylvain Teulier
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
- Department of Pulmonology and Thoracic Oncology, University Hospital of Caen, Caen, France
| | - Céline Bazille
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
- Department of Pathology, University Hospital of Caen, Caen, France
| | - Elise Letissier
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Jérôme Levallet
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Didier Divoux
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
| | - Méziane Ibazizène
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP CYCERON, 14000, Caen, France
| | - Stéphane Guillouet
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP CYCERON, 14000, Caen, France
| | - Cécile Perrio
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP CYCERON, 14000, Caen, France
| | - Louisa Barré
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP CYCERON, 14000, Caen, France
| | - Sébastien Serres
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Nicola R Sibson
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Françoise Chapon
- Department of Pathology, University Hospital of Caen, Caen, France
| | - Guénaëlle Levallet
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France
- Department of Pathology, University Hospital of Caen, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000, Caen, France.
| |
Collapse
|
96
|
Elming PB, Wittenborn TR, Busk M, Sørensen BS, Thomsen MBH, Strandgaard T, Dyrskjøt L, Nielsen S, Horsman MR. Refinement of an Established Procedure and Its Application for Identification of Hypoxia in Prostate Cancer Xenografts. Cancers (Basel) 2021; 13:2602. [PMID: 34073301 PMCID: PMC8198481 DOI: 10.3390/cancers13112602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This pre-clinical study was designed to refine a dissection method for validating the use of a 15-gene hypoxia classifier, which was previously established for head and neck squamous cell carcinoma (HNSCC) patients, to identify hypoxia in prostate cancer. METHODS PC3 and DU-145 adenocarcinoma cells, in vitro, were gassed with various oxygen concentrations (0-21%) for 24 h, followed by real-time PCR. Xenografts were established in vivo, and the mice were injected with the hypoxic markers [18F]-FAZA and pimonidazole. Subsequently, tumors were excised, frozen, cryo-sectioned, and analyzed using autoradiography ([18F]-FAZA) and immunohistochemistry (pimonidazole); the autoradiograms used as templates for laser capture microdissection of hypoxic and non-hypoxic areas, which were lysed, and real-time PCR was performed. RESULTS In vitro, all 15 genes were increasingly up-regulated as oxygen concentrations decreased. With the xenografts, all 15 genes were up-regulated in the hypoxic compared to non-hypoxic areas for both cell lines, although this effect was greater in the DU-145. CONCLUSIONS We have developed a combined autoradiographic/laser-guided microdissection method with broad applicability. Using this approach on fresh frozen tumor material, thereby minimizing the degree of RNA degradation, we showed that the 15-gene hypoxia gene classifier developed in HNSCC may be applicable for adenocarcinomas such as prostate cancer.
Collapse
Affiliation(s)
- Pernille B. Elming
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Thomas R. Wittenborn
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Morten Busk
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Brita S. Sørensen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Mathilde Borg Houlberg Thomsen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Steffen Nielsen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Michael R. Horsman
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| |
Collapse
|
97
|
Links between the unfolded protein response and the DNA damage response in hypoxia: a systematic review. Biochem Soc Trans 2021; 49:1251-1263. [PMID: 34003246 PMCID: PMC8286837 DOI: 10.1042/bst20200861] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Hypoxia is a feature of most solid tumours and predicts for poor prognosis. In radiobiological hypoxia (<0.1% O2) cells become up to three times more resistant to radiation. The biological response to radiobiological hypoxia is one of few physiologically relevant stresses that activates both the unfolded protein and DNA damage responses (UPR and DDR). Links between these pathways have been identified in studies carried out in normoxia. Based in part on these previous studies and recent work from our laboratory, we hypothesised that the biological response to hypoxia likely includes overlap between the DDR and UPR. While inhibition of the DDR is a recognised strategy for improving radiation response, the possibility of achieving this through targeting the UPR has not been realised. We carried out a systematic review to identify links between the DDR and UPR, in human cell lines exposed to <2% O2. Following PRISMA guidance, literature from January 2010 to October 2020 were retrieved via Ovid MEDLINE and evaluated. A total of 202 studies were included. LAMP3, ULK1, TRIB3, CHOP, NOXA, NORAD, SIAH1/2, DYRK2, HIPK2, CREB, NUPR1, JMJD2B, NRF2, GSK-3B, GADD45a, GADD45b, STAU1, C-SRC, HK2, CAV1, CypB, CLU, IGFBP-3 and SP1 were highlighted as potential links between the hypoxic DDR and UPR. Overall, we identified very few studies which demonstrate a molecular link between the DDR and UPR in hypoxia, however, it is clear that many of the molecules highlighted warrant further investigation under radiobiological hypoxia as these may include novel therapeutic targets to improve radiotherapy response.
Collapse
|
98
|
van Gisbergen MW, Zwilling E, Dubois LJ. Metabolic Rewiring in Radiation Oncology Toward Improving the Therapeutic Ratio. Front Oncol 2021; 11:653621. [PMID: 34041023 PMCID: PMC8143268 DOI: 10.3389/fonc.2021.653621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
To meet the anabolic demands of the proliferative potential of tumor cells, malignant cells tend to rewire their metabolic pathways. Although different types of malignant cells share this phenomenon, there is a large intracellular variability how these metabolic patterns are altered. Fortunately, differences in metabolic patterns between normal tissue and malignant cells can be exploited to increase the therapeutic ratio. Modulation of cellular metabolism to improve treatment outcome is an emerging field proposing a variety of promising strategies in primary tumor and metastatic lesion treatment. These strategies, capable of either sensitizing or protecting tissues, target either tumor or normal tissue and are often focused on modulating of tissue oxygenation, hypoxia-inducible factor (HIF) stabilization, glucose metabolism, mitochondrial function and the redox balance. Several compounds or therapies are still in under (pre-)clinical development, while others are already used in clinical practice. Here, we describe different strategies from bench to bedside to optimize the therapeutic ratio through modulation of the cellular metabolism. This review gives an overview of the current state on development and the mechanism of action of modulators affecting cellular metabolism with the aim to improve the radiotherapy response on tumors or to protect the normal tissue and therefore contribute to an improved therapeutic ratio.
Collapse
Affiliation(s)
- Marike W van Gisbergen
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Dermatology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Emma Zwilling
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
99
|
Zhao C, Zhou Y, Ma H, Wang J, Guo H, Liu H. A four-hypoxia-genes-based prognostic signature for oral squamous cell carcinoma. BMC Oral Health 2021; 21:232. [PMID: 33941139 PMCID: PMC8094530 DOI: 10.1186/s12903-021-01587-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/16/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the most common maligancies of the head and neck. The prognosis was is significantly different among OSCC patients. This study aims to identify new biomarkers to establish a prognostic model to predict the survival of OSCC patients. METHODS The mRNA expression and corresponding clinical information of OSCC patients were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus. Additionally, a total of 26 hypoxia-related genes were also obtained from a previous study. Univariate Cox regression analysis and LASSO Cox regression analysis were performed to screen the optimal hypoxia-related genes which were associated with the prognosis of OSCC. to establish the predictive model (Risk Score) was established for estimating the patient's overall survival (OS). Multivariate Cox regression analysis was used to determine whether the Risk Score was an independent prognostic factor. Based on all the independent prognostic factors, nomogram was established to predict the OS probability of OSCC patients. The relative proportion of 22 immune cell types in each patient was evaluated by CIBERSORT software. RESULTS We determined that a total of four hypoxia-related genes including ALDOA, P4HA1, PGK1 and VEGFA were significantly associated with the prognosis of OSCC patients. The nomogram established based on all the independent factors could reliably predict the long-term OS of OSCC patients. In addition, our resluts indicated that the inferior prognosis of OSCC patients with high Risk Score might be related to the immunosuppressive microenvironments. CONCLUSION This study shows that high expression of hypoxia-related genes including ALDOA, P4HA1, PGK1 and VEGFA is associated with poor prognosis in OSCC patients, and they can be used as potential markers for predicting prognosis in OSCC patients.
Collapse
Affiliation(s)
- Chenguang Zhao
- Department of Emergency and General Dentistry, Tianjin Stomatology Hospital, School of Medicine, NanKai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Yingrui Zhou
- Department of Emergency and General Dentistry, Tianjin Stomatology Hospital, School of Medicine, NanKai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Hongwei Ma
- Department of Emergency and General Dentistry, Tianjin Stomatology Hospital, School of Medicine, NanKai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Jinhui Wang
- Department of Emergency and General Dentistry, Tianjin Stomatology Hospital, School of Medicine, NanKai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Haoliang Guo
- Department of Emergency and General Dentistry, Tianjin Stomatology Hospital, School of Medicine, NanKai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Hao Liu
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatology Hospital, School of Medicine, NanKai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, No. 75, Dagu North Road, Heping District, Tianjin, 300041, China.
| |
Collapse
|
100
|
Mein S, Tessonnier T, Kopp B, Harrabi S, Abdollahi A, Debus J, Haberer T, Mairani A. Spot-Scanning Hadron Arc (SHArc) Therapy: A Study With Light and Heavy Ions. Adv Radiat Oncol 2021; 6:100661. [PMID: 33817410 PMCID: PMC8010580 DOI: 10.1016/j.adro.2021.100661] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/29/2020] [Accepted: 01/18/2021] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To evaluate the clinical potential of spot-scanning hadron arc (SHArc) therapy with a heavy-ion gantry. METHODS AND MATERIALS A series of in silico studies was conducted via treatment plan optimization in FRoG and the RayStation TPS to compare SHArc therapy against reference plans using conventional techniques with single, parallel-opposed, and 3-field configurations for 3 clinical particle beams (protons [p], helium [4He], and carbon [12C] ions). Tests were performed on water-equivalent cylindrical phantoms for simple targets and clinical-like scenarios with an organ-at-risk in proximity of the target. Effective dose and dose-averaged linear energy transfer (LETD) distributions for SHArc were evaluated against conventional planning techniques applying the modified microdosimetric kinetic model for considering bio-effect with (α/β)x = 2 Gy. A model for hypoxia-induced tumor radio-resistance was developed for particle therapy with dependence on oxygen concentration and particle species/energy (Zeff/β)2 to investigate the impact on effective dose. RESULTS SHArc plans exhibited similar target coverage with unique treatment attributes and distributions compared with conventional planning, with carbon ions demonstrating the greatest potential for tumor control and normal tissue sparing among the arc techniques. All SHArc plans exhibited a low-dose bath outside the target volume with a reduced maximum dose in normal tissues compared with single, parallel-opposed, and 3-field configuration plans. Moreover, favorable LETD distributions were made possible using the SHArc approach, with maximum LETD in the r = 5 mm tumor core (~8 keVμm-1, ~30 keVμm-1, and ~150 keVμm-1 for p, 4He, and 12C ions, respectively) and reductions of high-LET regions in normal tissues and organs-at-risk compared with static treatment beam delivery. CONCLUSION SHArc therapy offers potential treatment benefits such as increased normal tissue sparing. Without explicit consideration of oxygen concentration during treatment planning and optimization, SHArc-C may mitigate tumor hypoxia-induced loss of efficacy. Findings justify further development of robust SHArc treatment planning toward potential clinical translation.
Collapse
Affiliation(s)
- Stewart Mein
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Tessonnier
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Kopp
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Physics and Astronomy, Heidelberg University, Germany
| | - Semi Harrabi
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Physics and Astronomy, Heidelberg University, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Mairani
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- National Centre of Oncological Hadrontherapy (CNAO), Medical Physics, Pavia, Italy
| |
Collapse
|