1
|
Hill RM, Li C, Hughes JR, Rocha S, Grundy GJ, Parsons JL. Autophagy is the main driver of radioresistance of HNSCC cells in mild hypoxia. J Cell Mol Med 2024; 28:e18482. [PMID: 38899556 PMCID: PMC11187736 DOI: 10.1111/jcmm.18482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Hypoxia poses a significant challenge to the effectiveness of radiotherapy in head and neck squamous cell carcinoma (HNSCC) patients, and it is imperative to discover novel approaches to overcome this. In this study, we investigated the underlying mechanisms contributing to x-ray radioresistance in HPV-negative HNSCC cells under mild hypoxic conditions (1% oxygen) and explored the potential for autophagy modulation as a promising therapeutic strategy. Our findings show that HNSCC cells exposed to mild hypoxic conditions exhibit increased radioresistance, which is largely mediated by the hypoxia-inducible factor (HIF) pathway. We demonstrate that siRNA knockdown of HIF-1α and HIF-1β leads to increased radiosensitivity in HNSCC cells under hypoxia. Hypoxia-induced radioresistance was not attributed to differences in DNA double strand break repair kinetics, as these remain largely unchanged under normoxic and hypoxic conditions. Rather, we identify autophagy as a critical protective mechanism in HNSCC cells following irradiation under mild hypoxia conditions. Targeting key autophagy genes, such as BECLIN1 and BNIP3/3L, using siRNA sensitizes these cells to irradiation. Whilst autophagy's role in hypoxic radioresistance remains controversial, this study highlights the importance of autophagy modulation as a potential therapeutic approach to enhance the effectiveness of radiotherapy in HNSCC.
Collapse
Affiliation(s)
- Rhianna M. Hill
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Chun Li
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | - Jonathan R. Hughes
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | - Sonia Rocha
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Gabrielle J. Grundy
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Jason L. Parsons
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
2
|
Aggarwal N, Yadav J, Chhakara S, Janjua D, Tripathi T, Chaudhary A, Chhokar A, Thakur K, Singh T, Bharti AC. Phytochemicals as Potential Chemopreventive and Chemotherapeutic Agents for Emerging Human Papillomavirus-Driven Head and Neck Cancer: Current Evidence and Future Prospects. Front Pharmacol 2021; 12:699044. [PMID: 34354591 PMCID: PMC8329252 DOI: 10.3389/fphar.2021.699044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
Head and neck cancer (HNC) usually arises from squamous cells of the upper aerodigestive tract that line the mucosal surface in the head and neck region. In India, HNC is common in males, and it is the sixth most common cancer globally. Conventionally, HNC attributes to the use of alcohol or chewing tobacco. Over the past four decades, portions of human papillomavirus (HPV)-positive HNC are increasing at an alarming rate. Identification based on the etiological factors and molecular signatures demonstrates that these neoplastic lesions belong to a distinct category that differs in pathological characteristics and therapeutic response. Slow development in HNC therapeutics has resulted in a low 5-year survival rate in the last two decades. Interestingly, HPV-positive HNC has shown better outcomes following conservative treatments and immunotherapies. This raises demand to have a pre-therapy assessment of HPV status to decide the treatment strategy. Moreover, there is no HPV-specific treatment for HPV-positive HNC patients. Accumulating evidence suggests that phytochemicals are promising leads against HNC and show potential as adjuvants to chemoradiotherapy in HNC. However, only a few of these phytochemicals target HPV. The aim of the present article was to collate data on various leading phytochemicals that have shown promising results in the prevention and treatment of HNC in general and HPV-driven HNC. The review explores the possibility of using these leads against HPV-positive tumors as some of the signaling pathways are common. The review also addresses various challenges in the field that prevent their use in clinical settings.
Collapse
Affiliation(s)
- Nikita Aggarwal
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Joni Yadav
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Suhail Chhakara
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Divya Janjua
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Apoorva Chaudhary
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Arun Chhokar
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Kulbhushan Thakur
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Tejveer Singh
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Alok Chandra Bharti
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| |
Collapse
|
3
|
Ozturk S, Gorgun C, Gokalp S, Vatansever S, Sendemir A. Development and characterization of cancer stem cell-based tumoroids as an osteosarcoma model. Biotechnol Bioeng 2020; 117:2527-2539. [PMID: 32391924 DOI: 10.1002/bit.27381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/04/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
Abstract
Three-dimensional (3D) cancer tumor models are becoming vital approaches for high-throughput drug screening, drug targeting, development of novel theranostic systems, and personalized medicine. Yet, it is becoming more evident that the tumor progression and metastasis is fueled by a subpopulation of stem-like cells within the tumor that are also called cancer stem cells (CSCs). This study aimed to develop a tumoroid model using CSCs. For this purpose CD133+ cells were isolated from SaOS-2 osteosarcoma cell line with magnetic-activated cell sorting. To evaluate tumoroid formation ability, the cells were incubated in different cell numbers in agar gels produced by 3D Petri Dish® method. Subsequently, CD133+ cells and CD133- cells were co-cultured to investigate CD133+ cell localization in tumoroids. The characterization of tumoroids was performed using Live&Dead staining, immunohistochemistry, and quantitative polymerase chain reaction analysis. The results showed that, CD133+ , CD133- and SaOS-2 cells were all able to form 3D tumoroids regardless of the initial cell number, but, while 72 hr were needed for CD133+ cells to self-assemble, 24 hr were enough for CD133- and SaOS-2 cells. CD133+ cells were located within tumoroids randomly with high cell viability. Finally, when compared to two-dimensional (2D) cultures, there were 5.88, 4.14, 6.95, and 1.68-fold higher messenger RNA expressions for Sox2, OCT3/4, Nanog, and Nestin, respectively, in CD133+ cells that were cultured within 3D tumoroids, showing longer maintenance of stem cell phenotype in 3D, that can allow more relevant screening and targeting efficiency in pharmaceutical testing. It was concluded that CSC-based tumoroids are propitious as 3D tumor models to fill the gap between conventional 2D in vitro culture and in vivo animal experiments for cancer research.
Collapse
Affiliation(s)
- Sukru Ozturk
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.,Bioengineering Division, Institute for Graduate Studies in Science and Engineering, Hacettepe University, Ankara, Turkey.,Department of Biomedical Technologies, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Turkey
| | - Cansu Gorgun
- Department of Biomedical Technologies, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Turkey.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Sevtap Gokalp
- Department of Histology and Embryology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey.,Department of Histology and Embryology, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | - Seda Vatansever
- Department of Histology and Embryology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey.,Research Center of Experimental Health Sciences (DESAM), Near East University, Mersin, Turkey
| | - Aylin Sendemir
- Department of Biomedical Technologies, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Turkey.,Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey
| |
Collapse
|
4
|
Humtsoe JO, Pham E, Louie RJ, Chan DA, Kramer RH. ErbB3 upregulation by the HNSCC 3D microenvironment modulates cell survival and growth. Oncogene 2015; 35:1554-64. [PMID: 26073080 DOI: 10.1038/onc.2015.220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/06/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
Head and neck squamous carcinomas (HNSCC) present as dense epithelioid three-dimensional (3D) tumor nests that can mediate signals via the human epidermal growth factor receptor (ErbB) tyrosine kinase family to promote intratumoral survival and growth. We examined the role of the tumor microenvironment on ErbB receptor family expression and found that the status of intercellular organization altered the receptor profile. We showed that HNSCC cells forced into tumor island-like 3D aggregates strongly upregulated ErbB3 at the level of transcription. Not only was the elevated ErbB3 responsive to HRG-β1-induced enhanced signaling mechanism, but also analysis by siRNA-knockdown and kinase inhibitor strategies revealed that the ErbB3/AKT signaling pathway was sufficient to enhance tumor cell survival and growth potential. Elevated ErbB3 expression in the high-density 3D culture system was strongly associated with hypoxia-induced HIF-1α. Hypoxia-regulated ErbB3 expression was mediated by the HIF-1α-binding consensus sequence in the ErbB3 proximal promoter. The findings show that the local 3D tumor microenvironment can trigger reprograming and switching of ErbB family members and thereby influence ErbB3-driven tumor growth.
Collapse
Affiliation(s)
- J O Humtsoe
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - E Pham
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - R J Louie
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - D A Chan
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - R H Kramer
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Fu Z, Chen D, Cheng H, Wang F. Hypoxia-inducible factor-1α protects cervical carcinoma cells from apoptosis induced by radiation via modulation of vascular endothelial growth factor and p53 under hypoxia. Med Sci Monit 2015; 21:318-25. [PMID: 25623525 PMCID: PMC4316866 DOI: 10.12659/msm.893265] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background The aim of the study was to assess the role of irradiation in the expression of HIF-1α, VEGF, and P53 in human cervical carcinoma cells under a simulated hypoxia environment. Material/Methods The tetrazolium-based colorimetric cellular assay (MTT) and flow cytometry (FCM) were used to detect the growth inhibition rates of HeLa cells in different groups. Western blot and reverse transcription polymerase chain reaction (RT-PCR) were used to observe gene and protein expression of HIF-1α, VEGF, and P53. The effect of HIF-1α on radioresistance and expression of VEGF and P53 were confirmed with the HIF-1α siRNA in vivo and in vitro. Results Hypoxic conditions enhanced the radiation resistance dependent on HIF-1α by elevating the expression of VEGF and inhibiting the expression of p53. After transfection of HIF-1α siRNA, MTT assay showed the survival rates were increased in the cells receiving irradiation under hypoxia. The expression of VEGF decreased significantly more than that of cells transfected with sense oligodeoxynucleotides, while an opposite result was found in the expression of P53 protein in the same X-ray dose (p<0.05). In vivo, the radioresistance of HIF-1α was consistent with the results in vitro. Conclusions In the future, we might inhibit human cervical cancer progression and enhance the radiosensitivity by inhibiting HIF-1α to reduce VEGF and increase P53 expression. The challenge is how to use this information to optimize cancer therapy.
Collapse
Affiliation(s)
- Zhichao Fu
- Department of Radiology, Fuzhou General Hospital, Fuzhou, Fujian, China (mainland)
| | - Dongsheng Chen
- Department of Anesthesia, Fuzhou General Hospital, Fuzhou, Fujian, China (mainland)
| | - Huihua Cheng
- Department of Radiology, Fuzhou General Hospital, Fuzhou, Fujian, China (mainland)
| | - Fengmei Wang
- Department of Obstetrics and Gynecology, Fuzhou General Hospital, Fuzhou, Fujian, China (mainland)
| |
Collapse
|
6
|
Cellular Redox Status Regulates Emodin-Induced Radiosensitization of Nasopharyngeal Carcinoma Cells In Vitro and In Vivo. JOURNAL OF PHARMACEUTICS 2013; 2013:218297. [PMID: 26555969 PMCID: PMC4590808 DOI: 10.1155/2013/218297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/28/2013] [Accepted: 03/22/2013] [Indexed: 02/07/2023]
Abstract
Here, we report that regulation of cellular redox status is required for radiosensitization of nasopharyngeal carcinoma (NPC) cells by emodin. We evaluated emodin's radiosensitivity-enhancing ability by using NPC cells in vitro and xenografts in vivo. A clonogenic assay was performed to evaluate NPC cell survival and to determine dose modification factors. Flow cytometry, western blot analysis, and in vivo radiation-induced tumor regrowth delay assays were performed to characterize emodin's effects. Exposure of CNE-1 NPC cells to emodin enhanced their radiosensitivity. HIF-1α expression significantly increased under hypoxic conditions but did not change after treatment with emodin alone. Emodin downregulated mRNA and protein expression of HIF-1α. Cells exposed to radiation and emodin underwent significant cell cycle arrest at the G2/M phase. The percentage of apoptotic cells and reactive oxygen species (ROS) levels were significantly higher in the group exposed to emodin and radiation hypoxic group than in the other groups. Compared to the CNE-1 xenografts exposed to radiation alone, CNE-1 xenografts exposed to radiation with emodin showed significantly enhanced radiation effects. Our data suggest that emodin effectively enhanced the radiosensitivity of CNE-1 cells in vitro and in vivo. The mechanism appears to involve ROS generation and ROS-mediated inhibition of HIF-1α expression.
Collapse
|
7
|
Helbig L, Yaromina A, Sriramareddy SN, Böke S, Koi L, Thames HD, Baumann M, Zips D. Prognostic value of HIF-1α expression during fractionated irradiation. Strahlenther Onkol 2012; 188:1031-7. [PMID: 23053140 DOI: 10.1007/s00066-012-0150-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/18/2012] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE Hypoxia and reoxygenation are important determinants of outcome after radiotherapy. HIF-1α is a key molecule involved in cellular response to hypoxia. HIF-1α expression levels have been shown to change after irradiation. The objective of the present study was to explore the prognostic value of HIF-1α expression during fractionated irradiation. MATERIALS AND METHODS Six human squamous cell carcinoma models xenografted in nude mice were analysed. Tumours were excised after 3, 5 and 10 fractions. HIF-1α expression was quantified by western blot. For comparative analysis, previously published data on local tumour control data and pimonidazole hypoxic fraction was used. RESULTS HIF-1α expression in untreated tumours exhibited intertumoural heterogeneity and did not correlate with pimonidazole hypoxic fraction. During fractionated irradiation the majority of tumour models exhibited a decrease in HIF-1α expression, whereas in UT-SCC-5 no change was observed. Neither kinetics nor expression levels during fractionated irradiation correlated with local tumour control. CONCLUSION Our data do not support the use of HIF-1α determined during treatment as a biomarker to predict outcome after fractionated irradiation.
Collapse
Affiliation(s)
- L Helbig
- Dept. of Radiation Oncology/ OncoRay National Center for Radiation Research, Medical Faculty and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Przybyla BD, Shafirstein G, Koonce NA, Webber JS, Griffin RJ. Conductive thermal ablation of 4T1 murine breast carcinoma reduces severe hypoxia in surviving tumour. Int J Hyperthermia 2012; 28:156-62. [PMID: 22335229 DOI: 10.3109/02656736.2011.636783] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE The purpose of this study was to quantify hypoxia changes in viable tumour volumes after thermal ablation of a murine breast carcinoma. METHODS Murine breast 4T1 tumours were grown in the rear leg of BALB/c mice to an average diameter of 10-12 mm. Tumours were treated with conductive interstitial thermal therapy (CITT) at a peak temperature of 80-90°C for 10 min. The animals were euthanised 72 h later, and the tumours were removed for immunohistochemical staining with pimonidazole - a marker of partial pressure of oxygen. The levels of pimonidazole staining intensity were used to quantify changes in hypoxia gradients in terms of strong, medium and weak positive pixel fractions. RESULTS The pimonidazole staining ratio of viable control tumour tissue to viable tissue in tumours that were ablated was 0.7 for weak staining, 2.7 for medium staining and 8.0 (p < 0.03) for strong pimonidazole staining. CONCLUSION This shift of pimonidazole staining toward lower intensity pixels in the remaining tumour indicates that tumour ablation with CITT may increase radiosensitivity of the remaining tumour tissue and presents a rationale for combination therapy.
Collapse
Affiliation(s)
- Beata D Przybyla
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA.
| | | | | | | | | |
Collapse
|
9
|
Yang W, Sun T, Cao J, Fan S. Hypoxia-inducible factor-1α downregulation by small interfering RNA inhibits proliferation, induces apoptosis, and enhances radiosensitivity in chemical hypoxic human hepatoma SMMC-7721 cells. Cancer Biother Radiopharm 2011; 26:565-71. [PMID: 21950556 DOI: 10.1089/cbr.2011.0955] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Radiation plays an important role in the treatment of hepatoma. In order to improve its therapeutic ratio, there has been much interest in augmenting the effect of radiation on tumors by combining it with molecularly targeted therapeutics. Hypoxia-inducible factor-1 (HIF-1) is an excellent potential candidate for targeted molecular therapy to improve radiation outcome. In this study, HIF-1α-targeted small interfering RNA (siRNA) expression vector was constructed and transfected into human hepatoma SMMC-7721 cells, which followed by culture in CoCl(2)-induced hypoxia. HIF-1α downregulation by siRNA inhibited proliferation, induced apoptosis, and enhanced radiosensitivity in chemical hypoxic SMMC-7721 cells in vitro. These findings suggest that specific inhibition of HIF-1α expression in combination with radiotherapy would be expected to exert a strong antitumor effect on human hepatoma.
Collapse
Affiliation(s)
- Wei Yang
- Department of Radiobiology, School of Radiological Medicine and Public Health, Soochow University, No. 199 Renai Road, Suzhou, China.
| | | | | | | |
Collapse
|
10
|
See AP, Zeng J, Tran PT, Lim M. Acute toxicity of second generation HIV protease-inhibitors in combination with radiotherapy: a retrospective case series. Radiat Oncol 2011; 6:25. [PMID: 21414215 PMCID: PMC3064638 DOI: 10.1186/1748-717x-6-25] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 03/17/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There is little data on the safety of combining radiation therapy and human immunodeficiency virus (HIV) protease inhibitors to treat cancers in HIV-positive patients. We describe acute toxicities observed in a series of HIV-positive patients receiving modern radiation treatments, and compare patients receiving HIV protease inhibitors (PI) with patients not receiving HIV PIs. METHODS By reviewing the clinical records beginning January 1, 2009 from the radiation oncology department, we identified 29 HIV-positive patients who received radiation therapy to 34 body sites. Baseline information, treatment regimen, and toxicities were documented by review of medical records: patient age, histology and source of the primary tumor, HIV medication regimen, pre-radiation CD4 count, systemic chemotherapy, radiation therapy dose and fractionation, irradiated body region, toxicities, and duration of follow-up. Patients were grouped according to whether they received concurrent HIV PIs and compared using Pearson's chi-square test. RESULTS At baseline, the patients in the two groups were similar with the exception of HIV medication regimens, CD4 count and presence of AIDS-defining malignancy. Patients taking concurrent PIs were more likely to be taking other HIV medications (p = 0.001) and have CD4 count >500 (p = 0.006). Patients taking PIs were borderline less likely to have an AIDS-defining malignancy (p = 0.06). After radiation treatment, 100 acute toxicities were observed and were equally common in both groups (64 [median 3 per patient, IQR 1-7] with PIs; 36 [median 3 per patient, IQR 2-3] without PIs). The observed toxicities were also equally severe in the two groups (Grades I, II, III respectively: 30, 30, 4 with PIs; 23, 13, 0 without PIs: p = 0.38). There were two cases that were stopped early, one in each group; these were not attributable to toxicity. CONCLUSIONS In this study of recent radiotherapy in HIV-positive patients taking second generation PIs, no difference in toxicities was observed in patients taking PIs compared to patients not taking PIs during radiation therapy. This suggests that it is safe to use unmodified doses of PIs and radiation therapy in HIV cancer patients, and that it is feasible to use PIs as a radiosensitizer in cancer therapy, as has been suggested by pre-clinical results.
Collapse
Affiliation(s)
- Alfred P See
- Department of Radiation Oncology and Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21231, USA
| | | | | | | |
Collapse
|