1
|
Yang LN, Song YR, Zhang H, Tu HL, Lu MY, Liu DQ, Sui JD, Li D, Xie Y, Wang Y. Monocyte count combined with GTVnx is an independent prognostic factor in non-metastatic nasopharyngeal carcinoma receiving radiotherapy. Front Oncol 2025; 15:1541212. [PMID: 40265015 PMCID: PMC12011717 DOI: 10.3389/fonc.2025.1541212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background and purpose The relationship between peripheral blood monocyte count and primary gross tumor volume with survival prognosis in newly diagnosed nasopharyngeal carcinoma(NPC) patients who received radiotherapy remains unclear. Therefore, We conducted a cohort study to assess the association of peripheral blood monocyte count and primary gross tumor volume with survival outcomes in newly diagnosed non-metastatic NPC patients who received radiotherapy. Materials/methods We included newly diagnosed non-metastatic NPC patients who underwent radiotherapy in our hospital from January 2013 to December 2015. General clinical characteristics such as age, gender, ECOG score and tumor stage, peripheral blood monocyte count, lymphocyte count, white blood cell count (WBC), neutrophil count, radiotherapy technology, total radiotherapy days, gross tumor volume of nasopharyngeal carcinoma (GTVnx) and gross tumor volume of cervix node (GTVnd) of patients before radiotherapy, and whether chemotherapy was induced were recorded. The primary endpoint was overall survival, the secondary endpoint was progression-free survival. Univariate and multivariate COX regression were used to analyze the relationship among peripheral blood monocyte count, GTVnx, and survival outcomes. Spearman correlation analysis was used to analyze the correlation between risk factors. Based on the independent risk factors for OS, we further divide patients into three different risk groups, and the differences in clinical and therapeutic indicators and survival outcomes between the three groups were analyzed using a one-way analysis of variance. Results A total of 448 participants were included in the study, the median follow-up time was 74.3 months. Of these, 97 (21.7%) died. In the univariate and multivariate Cox regression analyses, peripheral blood monocyte count and GTVnx were independently associated with OS. The high monocyte count and GTVnx were associated with the poor OS and PFS. Correlation analysis showed that monocyte count was positively correlated with WBC, platelet, and neutrophil. GTVnx was positively correlated with platelet, neutrophil, and Epstein-Barr virus before treatment. Survival curves significantly differed among patients in different risk groups for OS (p = 0.0008) and PFS (p = 0.0007). Besides, For every increase in monocyte unit count, the OS and PFS risks of patients in the low GTVnx group increased by 2.64 and 2.31 folds, respectively. Conclusions Peripheral blood monocyte count combined with GTVnx is an independent predictor for overall survival and progression free-survival in newly diagnosed non-metastatic NPC patients who received radiotherapy. The benefit of patients with GTVnx< 28.5cm3 could be remarkably attenuated by the high monocyte count.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dan Li
- Radiation Oncology Center, Chongqing University Cancer Hospital, School of Medicine,
Chongqing University, Chongqing, China
| | - Yue Xie
- Radiation Oncology Center, Chongqing University Cancer Hospital, School of Medicine,
Chongqing University, Chongqing, China
| | - Ying Wang
- Radiation Oncology Center, Chongqing University Cancer Hospital, School of Medicine,
Chongqing University, Chongqing, China
| |
Collapse
|
2
|
You P, Liu S, Li Q, Xie D, Yao L, Guo C, Guo Z, Wang T, Qiu H, Guo Y, Li J, Zhou H. Radiation-sensitive genetic prognostic model identifies individuals at risk for radiation resistance in head and neck squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:15623-15640. [PMID: 37656244 DOI: 10.1007/s00432-023-05304-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/15/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND The advantages of radiotherapy for head and neck squamous cell carcinoma (HNSCC) depend on the radiation sensitivity of the patient. Here, we established and verified radiological factor-related gene signature and built a prognostic risk model to predict whether radiotherapy would be beneficial. METHODS Data from The Cancer Genome Atlas, Gene Expression Omnibus, and RadAtlas databases were subjected to LASSO regression, univariate COX regression, and multivariate COX regression analyses to integrate genomic and clinical information from patients with HNSCC. HNSCC radiation-related prognostic genes were identified, and patients classified into high- and low-risk groups, based on risk scores. Variations in radiation sensitivity according to immunological microenvironment, functional pathways, and immunotherapy response were investigated. Finally, the expression of HNSCC radiation-related genes was verified by qRT-PCR. RESULTS We built a clinical risk prediction model comprising a 15-gene signature and used it to divide patients into two groups based on their susceptibility to radiation: radiation-sensitive and radiation-resistant. Overall survival was significantly greater in the radiation-sensitive than the radiation-resistant group. Further, our model was an independent predictor of radiotherapy response, outperforming other clinical parameters, and could be combined with tumor mutational burden, to identify the target population with good predictive value for prognosis at 1, 2, and 3 years. Additionally, the radiation-resistant group was more vulnerable to low levels of immune infiltration, which are significantly associated with DNA damage repair, hypoxia, and cell cycle regulation. Tumor Immune Dysfunction and Exclusion scores also suggested that the resistant group would respond less favorably to immunotherapy. CONCLUSIONS Our prognostic model based on a radiation-related gene signature has potential for application as a tool for risk stratification of radiation therapy for patients with HNSCC, helping to identify candidates for radiation therapy and overcome radiation resistance.
Collapse
Affiliation(s)
- Peimeng You
- Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - Shengbo Liu
- Second Clinical College of Medicine, Southern Medical University, Guangzhou, China
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiaxuan Li
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Daipeng Xie
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangzhou, China
| | - Lintong Yao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Chenguang Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zefeng Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ting Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongrui Qiu
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yangzhong Guo
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - Junyu Li
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China.
| | - Haiyu Zhou
- Nanchang University, Nanchang, China.
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Jiangxi Lung Cancer Institute, Nanchang, China.
| |
Collapse
|
3
|
Flournoy J, Ashkanani S, Chen Y. Mechanical regulation of signal transduction in angiogenesis. Front Cell Dev Biol 2022; 10:933474. [PMID: 36081909 PMCID: PMC9447863 DOI: 10.3389/fcell.2022.933474] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/28/2022] [Indexed: 11/21/2022] Open
Abstract
Biophysical and biochemical cues work in concert to regulate angiogenesis. These cues guide angiogenesis during development and wound healing. Abnormal cues contribute to pathological angiogenesis during tumor progression. In this review, we summarize the known signaling pathways involved in mechanotransduction important to angiogenesis. We discuss how variation in the mechanical microenvironment, in terms of stiffness, ligand availability, and topography, can modulate the angiogenesis process. We also present an integrated view on how mechanical perturbations, such as stretching and fluid shearing, alter angiogenesis-related signal transduction acutely, leading to downstream gene expression. Tissue engineering-based approaches to study angiogenesis are reviewed too. Future directions to aid the efforts in unveiling the comprehensive picture of angiogenesis are proposed.
Collapse
Affiliation(s)
- Jennifer Flournoy
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, United States
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, United States
| | - Shahad Ashkanani
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, United States
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, United States
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
4
|
Niu K, Chen XW, Qin Y, Zhang LP, Liao RX, Sun JG. Celecoxib Blocks Vasculogenic Mimicry via an Off-Target Effect to Radiosensitize Lung Cancer Cells: An Experimental Study. Front Oncol 2021; 11:697227. [PMID: 34568026 PMCID: PMC8461170 DOI: 10.3389/fonc.2021.697227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/18/2021] [Indexed: 12/29/2022] Open
Abstract
The resistance to radiotherapy in lung cancer can be attributed to vasculogenic mimicry (VM) to some extent. Celecoxib (CXB), a selective inhibitor of cyclooxygenase-2 (COX-2), is reported as a radiosensitizer in non-small cell lung cancer (NSCLC). However, whether CXB can regulate VM formation via an off-target effect to radiosensitize NSCLC remains unclear. This study aimed to elucidate the mechanism underlying the radiosensitizing effect of CXB on NSCLC, i.e., whether CXB can inhibit VM formation via binding to newly identified targets other than COX-2. CXB radiosensitivity assay was performed in BALB/c mice bearing H460 xenografts and C57 mice bearing Lewis lung cancer (LLC) xenografts, which were divided into the control, CXB, irradiation (IR) treatment, and IR plus CXB groups. VM formation was observed using 3D Matrigel, periodic acid solution (PAS) staining, and immunofluorescence staining. The potential off-targets of CXB were screened using Protein Data Bank (PDB) database, MGLTools 1.5.6, and AutoDock Vina 1.1.2 and confirmed by Western blotting, enzyme activity assay, and RNA interference in vitro experiments and by immunohistochemistry in vivo experiments. CXB treatment almost eliminated the enhancement of VM formation by IR in vitro and in vivo, partially due to COX-2 inhibition. Four potential off-targets were predicted by molecular docking. Among them, aminopeptidase N (APN) and integrin alpha-V (ITAV) were remarkably inhibited in protein expression and enzyme activity in vitro or in vivo, consistent with the remarkable reduction of VM formation in H460 xenografts in BALB/c mice. In conclusion, CXB dramatically blocked VM through inhibiting newly identified off-targets APN and ITAV, other than COX-2, then radiosensitizing NSCLC.
Collapse
Affiliation(s)
- Kai Niu
- Cancer Institute of Chinese People's Liberation Army (PLA), Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xie-Wan Chen
- Cancer Institute of Chinese People's Liberation Army (PLA), Xinqiao Hospital, Army Medical University, Chongqing, China.,Medical English Department, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Yu Qin
- Nutrition and Food Hygiene Department, Institute of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Lu-Ping Zhang
- Cancer Institute of Chinese People's Liberation Army (PLA), Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Rong-Xia Liao
- Medical English Department, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Jian-Guo Sun
- Cancer Institute of Chinese People's Liberation Army (PLA), Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
5
|
Chua KLM, Chu PL, Tng DJH, Soo KC, Chua MLK. Repurposing Proton Beam Therapy through Novel Insights into Tumour Radioresistance. Clin Oncol (R Coll Radiol) 2021; 33:e469-e481. [PMID: 34509347 DOI: 10.1016/j.clon.2021.08.013] [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: 05/05/2021] [Revised: 08/02/2021] [Accepted: 08/25/2021] [Indexed: 12/11/2022]
Abstract
Despite improvements in radiotherapy, radioresistance remains an important clinical challenge. Radioresistance can be mediated through enhanced DNA damage response mechanisms within the tumour or through selective pressures exerted by the tumour microenvironment (TME). The effects of the TME have in recent times gained increased attention, in part due to the success of immune modulating strategies, but also through improved understanding of the downstream effects of hypoxia and dysregulated wound healing processes on mediating radioresistance. Although we have a better appreciation of these molecular mechanisms, efforts to address them through novel combination approaches have been scarce, owing to limitations of photon therapy and concerns over toxicity. At the same time, proton beam therapy (PBT) represents an advancement in radiotherapy technologies. However, early clinical results have been mixed and the clinical strategies around optimal use and patient selection for PBT remain unclear. Here we highlight the role that PBT can play in addressing radioresistance, through better patient selection, and by providing an improved toxicity profile for integration with novel agents. We will also describe the developments around FLASH PBT. Through close examination of its normal tissue-sparing effects, we will highlight how FLASH PBT can facilitate combination strategies to tackle radioresistance by further improving toxicity profiles and by directly mediating the mechanisms of radioresistance.
Collapse
Affiliation(s)
- K L M Chua
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - P L Chu
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore
| | - D J H Tng
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - K C Soo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore; Division of Surgical Oncology, National Cancer Centre Singapore, Singapore
| | - M L K Chua
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore; Division of Medical Sciences, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
6
|
Sheikholeslami S, Aryafar T, Abedi-Firouzjah R, Banaei A, Dorri-Giv M, Zamani H, Ataei G, Majdaeen M, Farhood B. The role of melatonin on radiation-induced pneumonitis and lung fibrosis: A systematic review. Life Sci 2021; 281:119721. [PMID: 34146555 DOI: 10.1016/j.lfs.2021.119721] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/19/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Pneumonitis and lung fibrosis, as the most common compliances of lung irradiation, can affect the quality of life. The use of radio-protective agents can ameliorate these injuries. This study aimed to review the potential protective role of melatonin in the treatment of radiation-induced Pneumonitis and lung fibrosis. METHODS The current systematic study was conducted based on PRISMA guidelines to identify relevant literature on " the effect of melatonin on radiation-induced pneumonitis and lung fibrosis" in the electronic databases of Web of Science, Embase, PubMed, and Scopus up to January 2021. Eighty-one articles were screened in accordance with the inclusion and exclusion criteria of the study. Finally, eight articles were included in this systematic review. RESULTS The finding showed that the lung irradiation-induced pneumonitis and lung fibrosis. The co-treatment with melatonin could alleviate these compliances through its anti-oxidant and anti-inflammatory actions. Melatonin through upregulation of some enzymes such as catalase, superoxide dismutase, glutathione, NADPH oxidases 2 and 4, dual oxidases 1 and 2, and also downregulation of malondialdehyde reduced oxidative stress following lung radiation. Moreover, melatonin through its anti-inflammatory effects, can attenuate the increased levels of nuclear factor kappa B, tumor necrosis factor alpha, transforming growth factor beta 1, SMAD2, interleukin (IL)-4, IL-4 receptor-a1 (IL4ra1), and IL-1 beta following lung radiation. The histological damages induced by ionizing radiation were also alleviated by co-treatment with melatonin. CONCLUSION According to the obtained results, it was found that melatonin can have anti-pneumonitis and anti-fibrotic following lung irradiation.
Collapse
Affiliation(s)
- Sahar Sheikholeslami
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Tayebeh Aryafar
- Department of Radiation Sciences, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Amin Banaei
- Department of Medical Physics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoumeh Dorri-Giv
- Nuclear Medicine Research Center, Department of Nuclear Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Zamani
- Department of Medical Physics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Gholamreza Ataei
- Department of Radiology Technology, Faculty of Paramedical Sciences, Babol University of Medical Science, Babol, Iran
| | - Mehrsa Majdaeen
- Department of Radiotherapy and Oncology, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
7
|
Im JH, Buzzelli JN, Jones K, Franchini F, Gordon-Weeks A, Markelc B, Chen J, Kim J, Cao Y, Muschel RJ. FGF2 alters macrophage polarization, tumour immunity and growth and can be targeted during radiotherapy. Nat Commun 2020; 11:4064. [PMID: 32792542 PMCID: PMC7426415 DOI: 10.1038/s41467-020-17914-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Regulation of the programming of tumour-associated macrophages (TAMs) controls tumour growth and anti-tumour immunity. We examined the role of FGF2 in that regulation. Tumours in mice genetically deficient in low-molecular weight FGF2 (FGF2LMW) regress dependent on T cells. Yet, TAMS not T cells express FGF receptors. Bone marrow derived-macrophages from Fgf2LMW−/− mice co-injected with cancer cells reduce tumour growth and express more inflammatory cytokines. FGF2 is induced in the tumour microenvironment following fractionated radiation in murine tumours consistent with clinical reports. Combination treatment of in vivo tumours with fractionated radiation and a blocking antibody to FGF2 prolongs tumour growth delay, increases long-term survival and leads to a higher iNOS+/CD206+ TAM ratio compared to irradiation alone. These studies show for the first time that FGF2 affects macrophage programming and is a critical regulator of immunity in the tumour microenvironment. Macrophages contribute to tumour progression and response to therapy. Here, the authors show that absence of FGF2 in the tumour microenvironment reduces tumour growth and enhances the anti-tumour immune response by altering macrophage polarization. As a result, disruption of this macrophage programming by anti-FGF2 blocking antibodies enhances the outcome from radiotherapy.
Collapse
Affiliation(s)
- Jae Hong Im
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jon N Buzzelli
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Keaton Jones
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Fanny Franchini
- The Kennedy Institute of Rheumatology, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Bostjan Markelc
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jianzhou Chen
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jin Kim
- Galaxy Biotech, 1230 Bordeaux Dr, Sunnyvale, CA, 94089, USA
| | - Yunhong Cao
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Ruth J Muschel
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
8
|
Farhood B, Ghorbani M, Abdi Goushbolagh N, Najafi M, Geraily G. Different Methods of Measuring Neutron Dose/Fluence Generated During Radiation Therapy with Megavoltage Beams. HEALTH PHYSICS 2020; 118:65-74. [PMID: 31764421 DOI: 10.1097/hp.0000000000001130] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Medical linear accelerators (linacs) are the most frequently applied radiation therapy machines in the locoregional treatment of cancers by producing either high-energy electron or photon beams. However, with high-energy photons (>8 MeV), interaction of these photons with different high-Z nuclei of materials in components of the linac head unavoidably generates neutrons. On the other hand, the average energy of these generated neutrons has almost the highest radiation-weighting factor. Therefore, the produced neutrons should not be neglected. There are various tools for the measurement of neutron dose/fluence generated in a megavoltage linac, including thermoluminescent dosimeters, solid-state nuclear track detectors, bubble detectors, activation foils, Bonner sphere systems, and ionization chamber pairs. In this review article, each of the above-mentioned dosimetric methods will be described in detail.
Collapse
Affiliation(s)
- Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehdi Ghorbani
- Biomedical Engineering and Medical Physics Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nouraddin Abdi Goushbolagh
- Medical Physics Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazale Geraily
- Medical Physics and Medical Engineering Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Mahdavi SR, Tutuni M, Farhood B, Nafisi N, Ghasemi S, Mirzaee H, Ahmadi S, Alizadeh A. Measurement of peripheral dose to the pelvic region and the associated risk for cancer development after breast intraoperative electron radiation therapy. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2019; 39:278-291. [PMID: 30634170 DOI: 10.1088/1361-6498/aafdc8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This study aimed to measure the received dose to the pelvic region of patients during breast intraoperative electron radiation therapy (IOERT). Furthermore, we compared the findings with those of external beam radiation therapy. Finally, secondary ovarian and uterus cancer risks following breast IOERT were estimated. In the current study, the received dose to the pelvic surface of 18 female patients during breast IOERT boosts were measured by thermoluminescent dosimeter (TLD-100) chips. All patients were treated with 12 Gy given in a single fraction. To estimate the dose to the ovary and uterus of the patients, conversion coefficients for depth from the surface dose were obtained in a Rando phantom. Given the received dose to the pelvic region of the patients, secondary ovarian and uterus cancer risks following breast IOERT were estimated. The received doses to the ovary and uterus surface of the patients were 0.260 ± 0.155 mGy to 31.460 ± 6.020 mGy and 0.485 ± 0.122 mGy to 22.387 ± 15.476 mGy, respectively. Corresponding intra-pelvic (ovary and uterus) regional doses were 0.012 ± 0.007 mGy to 1.479 ± 0.283 mGy and 0.027 ± 0.001 mGy to 1.164 ± 0.805 mGy, respectively. Findings demonstrated that the ratio of the received dose by the pelvic surface to the regional dose during breast IOERT was much less than external beam radiation therapy. The mean of the secondary cancer risks for the ovary in 8 and 10 MeV electron beam energies were 135.722 ± 117.331 × 10-6 and 69.958 ± 28.072 × 10-6, and for the uterus were 17.342 ± 10.583 × 10-6 and 2.971 ± 3.604 × 10-6, respectively. According to our findings, the use of breast IOERT in pregnant patients can be considered as a safe radiotherapeutic technique, because the received dose to the fetus was lower than 50 mGy. Furthermore, IOERT can efficiently reduce the unnecessary dose to the pelvic region and lowers the risk of secondary ovarian and uterus cancer following breast irradiation.
Collapse
Affiliation(s)
- Seyed Rabie Mahdavi
- Department of Medical Physics, Iran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Schernberg A, Blanchard P, Chargari C, Ou D, Levy A, Gorphe P, Breuskin I, Atallah S, Caula A, Escande A, Janot F, Nguyen F, Temam S, Deutsch E, Tao Y. Leukocytosis, prognosis biomarker in locally advanced head and neck cancer patients after chemoradiotherapy. Clin Transl Radiat Oncol 2018; 12:8-15. [PMID: 30073209 PMCID: PMC6068078 DOI: 10.1016/j.ctro.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/14/2018] [Accepted: 07/04/2018] [Indexed: 12/30/2022] Open
Abstract
Objective To study the prognostic value of leukocyte increase in a retrospective cohort of locally advanced head and neck squamous cell carcinoma (HNSCC) patients receiving definitive concurrent cisplatin and radiation. Materials and methods Clinical records of consecutive previously untreated locally advanced HNSCC patients treated in our Institution between March 2006 and October 2012 by concurrent cisplatin (100 mg/m2, every 3 weeks) and radiation (70 Gy in 7 weeks) were collected. The prognostic value of pretreatment leukocyte increase was examined, with focus on patterns of relapse and survival. Leukocytosis and neutrophilia were defined as a leukocyte count or a neutrophils count exceeding 10 and 7.5 G/L, respectively. Results We identified 193 patients, all treated with concurrent cisplatin-based chemoradiotherapy. Respectively 24% and 20% patients displayed baseline leukocytosis or neutrophilia. Mean leukocyte count were significantly more elevated in current smokers, patients with performance status (PS) >0, T4 and less in HPV + tumor. The 5-year actuarial overall survival (OS) and progression-free survival (PFS) were 56% and 51% respectively. In univariate analysis, both leukocytosis and neutrophilia were strongly associated with worse OS and PFS (p < 0.001). In multivariate analysis, N classification, HPV/p16, smoking status and leukocytosis were associated with worse OS and PFS. Patients with <3 cycles of cisplatin had worse survival. Conclusion In locally advanced HNSCC treated with concurrent cisplatin and radiation, baseline leukocytosis predicts OS and PFS. In addition with HPV status, this independent biomarker could help identifying patients with high risk of tumor relapse.
Collapse
Affiliation(s)
- Antoine Schernberg
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Pierre Blanchard
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Cyrus Chargari
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France.,INSERM1030, Gustave Roussy Cancer Campus, Villejuif France.,French Military Health Services Academy, Ecole du Val-de-Grâce, Paris, France.,Institut de Recherche Biomédicale des Armées, Bretigny-sur-Orge, France
| | - Dan Ou
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antonin Levy
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France.,INSERM1030, Gustave Roussy Cancer Campus, Villejuif France
| | - Philippe Gorphe
- Head and Neck Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ingrid Breuskin
- Head and Neck Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sarah Atallah
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alexandre Caula
- Head and Neck Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alexandre Escande
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - François Janot
- Head and Neck Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - France Nguyen
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Stéphane Temam
- Head and Neck Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France.,INSERM1030, Gustave Roussy Cancer Campus, Villejuif France.,Université Paris Sud, Université Paris Saclay, Faculté de médecine du Kremlin-Bicetre, Le Kremlin-Bicetre, France
| | - Yungan Tao
- Radiation Oncology Department, Gustave Roussy Cancer Campus, Villejuif, France.,INSERM1030, Gustave Roussy Cancer Campus, Villejuif France
| |
Collapse
|
11
|
McBride WH, Ganapathy E, Lee MH, Nesseler JP, Nguyen C, Schaue D. A perspective on the impact of radiation therapy on the immune rheostat. Br J Radiol 2017; 90:20170272. [PMID: 28707537 PMCID: PMC5853348 DOI: 10.1259/bjr.20170272] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The advent and success of immune checkpoint inhibitors (ICIs) in cancer treatment has broadened the spectrum of tumours that might be considered "immunogenic" and susceptible to immunotherapeutic (IT) intervention. Not all cancer types are sensitive, and not all patients with any given type respond. Combination treatment of ICIs with an established cytotoxic modality such as radiation therapy (RT) is a logical step towards improvement. For one, RT alone has been shown to be genuinely immunomodulatory and secondly pre-clinical data generally support combined ICI-RT approaches. This new integrated therapy for cancer treatment holds much promise, although there is still a lot to be learned about how best to schedule the treatments, manage the toxicities and determine what biomarkers might predict response, as well as many other issues. This review examines how RT alters the immune rheostat and how it might best be positioned to fully exploit IT.
Collapse
Affiliation(s)
- William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ekambaram Ganapathy
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Mi-Heon Lee
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jean P Nesseler
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Christine Nguyen
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
12
|
Jeong H, Bok S, Hong BJ, Choi HS, Ahn GO. Radiation-induced immune responses: mechanisms and therapeutic perspectives. Blood Res 2016; 51:157-163. [PMID: 27722125 PMCID: PMC5054246 DOI: 10.5045/br.2016.51.3.157] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/13/2016] [Accepted: 09/13/2016] [Indexed: 01/22/2023] Open
Abstract
Recent advancement in the radiotherapy technology has allowed conformal delivery of high doses of ionizing radiation precisely to the tumors while sparing large volume of the normal tissues, which have led to better clinical responses. Despite this technological advancement many advanced tumors often recur and they do so within the previously irradiated regions. How could tumors recur after receiving such high ablative doses of radiation? In this review, we outlined how radiation can elicit anti-tumor responses by introducing some of the cytokines that can be induced by ionizing radiation. We then discuss how tumor hypoxia, a major limiting factor responsible for failure of radiotherapy, may also negatively impact the anti-tumor responses. In addition, we highlight how there may be other populations of immune cells including regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs) that can be recruited to tumors interfering with the anti-tumor immunity. Finally, the impact of irradiation on tumor hypoxia and the immune responses according to different radiotherapy regimen is also delineated. It is indeed an exciting time to see that radiotherapy is being combined with immunotherapy in the clinic and we hope that this review can add an excitement to the field.
Collapse
Affiliation(s)
- Hoibin Jeong
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Seoyeon Bok
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Beom-Ju Hong
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Hyung-Seok Choi
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - G-One Ahn
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| |
Collapse
|
13
|
Giuliani M, Sampson LR, Wong O, Gay J, Le LW, Cho BCJ, Brade A, Sun A, Bezjak A, Hope AJ. Prognostic value of pretreatment circulating neutrophils, monocytes, and lymphocytes on outcomes in lung stereotactic body radiotherapy. ACTA ACUST UNITED AC 2016; 23:e362-8. [PMID: 27536185 DOI: 10.3747/co.23.3051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE In the present study, we determined the association of pretreatment circulating neutrophils, monocytes, and lymphocytes with clinical outcomes after lung stereotactic body radiotherapy (sbrt). METHODS All patients with primary lung cancer and with a complete blood count within 3 months of lung sbrt from 2005 to 2012 were included. Overall survival (os) was calculated using the Kaplan-Meier method. Factors associated with os were investigated using univariable and multivariable Cox proportional hazards regression. Fine-Gray competing risk regression was performed to test the association of the neutrophil:lymphocyte (nlr) and monocyte:lymphocyte (mlr) ratios with two types of failure: disease-related failure and death, and death unrelated to disease. RESULTS Of the 299 sbrt patients identified, 122 were eligible for analysis. The median and range of the nlr and mlr were 3.0 (0.3-22.0) and 0.4 (0.1-1.9) respectively. On multivariable analysis, sex (p = 0.02), T stage (p = 0.04), and nlr (p < 0.01) were associated with os. On multivariable analysis, T stage (p < 0.01) and mlr (p < 0.01) were associated with disease-related failure; mlr (p = 0.03), nlr (p < 0.01), and sbrt dose of 48 Gy in 4 fractions (p = 0.03) and 54 Gy or 60 Gy in 3 fractions (p = 0.02) were associated with disease-unrelated death. Median survival was 4.3 years in the nlr≤3 group (95% confidence interval: 3.5 to not reached) and 2.5 years in the nlr>3 group (95% confidence interval: 1.7 to 4.8; p < 0.01). CONCLUSIONS In lung sbrt patients, nlr and mlr are independently associated with os and disease-unrelated death. If validated, nlr and mlr could help to identify patients who would benefit most from sbrt.
Collapse
Affiliation(s)
- M Giuliani
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - L R Sampson
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - O Wong
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - J Gay
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | | | - B C J Cho
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - A Brade
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - A Sun
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - A Bezjak
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - A J Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON
| |
Collapse
|
14
|
Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 2015; 15:409-25. [PMID: 26105538 PMCID: PMC4896389 DOI: 10.1038/nrc3958] [Citation(s) in RCA: 1492] [Impact Index Per Article: 149.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Radiotherapy plays a central part in curing cancer. For decades, most research on improving treatment outcomes has focused on modulating radiation-induced biological effects on cancer cells. Recently, we have better understood that components within the tumour microenvironment have pivotal roles in determining treatment outcomes. In this Review, we describe vascular, stromal and immunological changes that are induced in the tumour microenvironment by irradiation and discuss how these changes may promote radioresistance and tumour recurrence. We also highlight how this knowledge is guiding the development of new treatment paradigms in which biologically targeted agents will be combined with radiotherapy.
Collapse
Affiliation(s)
- Holly E. Barker
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| | - James T. E. Paget
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Aadil A. Khan
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Kevin J. Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, SW3 6JB, UK
| |
Collapse
|
15
|
Schaue D, McBride WH. Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol 2015; 12:527-40. [PMID: 26122185 DOI: 10.1038/nrclinonc.2015.120] [Citation(s) in RCA: 497] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The past 20 years have seen dramatic changes in the delivery of radiation therapy, but the impact of radiobiology on the clinic has been far less substantial. A major consideration in the use of radiotherapy has been on how best to exploit differences between the tumour and host tissue characteristics, which in the past has been achieved empirically by radiation-dose fractionation. New advances are uncovering some of the mechanistic processes that underlie this success story. In this Review, we focus on how these processes might be targeted to improve the outcome of radiotherapy at the individual patient level. This approach would seem a more productive avenue of treatment than simply trying to increase the radiation dose delivered to the tumour.
Collapse
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| | - William H McBride
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| |
Collapse
|
16
|
Finkelstein SE, Salenius S, Mantz CA, Shore ND, Fernandez EB, Shulman J, Myslicki FA, Agassi AM, Rotterman Y, DeVries T, Sims R. Combining Immunotherapy and Radiation for Prostate Cancer. Clin Genitourin Cancer 2015; 13:1-9. [DOI: 10.1016/j.clgc.2014.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/29/2014] [Accepted: 09/17/2014] [Indexed: 02/06/2023]
|
17
|
Huang SH, Waldron JN, Milosevic M, Shen X, Ringash J, Su J, Tong L, Perez-Ordonez B, Weinreb I, Bayley AJ, Kim J, Hope A, Cho BJ, Giuliani M, Razak A, Goldstein D, Shi W, Liu FF, Xu W, O'Sullivan B. Prognostic value of pretreatment circulating neutrophils, monocytes, and lymphocytes in oropharyngeal cancer stratified by human papillomavirus status. Cancer 2014; 121:545-55. [DOI: 10.1002/cncr.29100] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/05/2014] [Accepted: 09/17/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Shao Hui Huang
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - John N. Waldron
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
- Department of Otolaryngology-Head & Neck Surgery; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Michael Milosevic
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Xiaowei Shen
- Department of Biostatistics; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Jolie Ringash
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Jie Su
- Department of Biostatistics; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Li Tong
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Bayardo Perez-Ordonez
- Department of Pathology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Ilan Weinreb
- Department of Pathology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Andrew J. Bayley
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - John Kim
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Andrew Hope
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - B.C. John Cho
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Meredith Giuliani
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Albiruni Razak
- Division of Medical Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - David Goldstein
- Department of Otolaryngology-Head & Neck Surgery; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Willa Shi
- Ontario Cancer Institute; University Health Network; Toronto Canada
| | - Fei-Fei Liu
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
- Ontario Cancer Institute; University Health Network; Toronto Canada
| | - Wei Xu
- Department of Biostatistics; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
| | - Brian O'Sullivan
- Department of Radiation Oncology; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
- Department of Otolaryngology-Head & Neck Surgery; Princess Margaret Cancer Centre/University of Toronto; Toronto Canada
- Ontario Cancer Institute; University Health Network; Toronto Canada
| |
Collapse
|
18
|
Boss MK, Bristow R, Dewhirst MW. Linking the history of radiation biology to the hallmarks of cancer. Radiat Res 2014; 181:561-77. [PMID: 24811865 PMCID: PMC4072211 DOI: 10.1667/rr13675.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hanahan and Weinberg recently updated their conceptual framework of the "Hallmarks of Cancer". The original article, published in 2000, is among the most highly cited reviews in the field of oncology. The goal of this review is to highlight important discoveries in radiation biology that pertain to the Hallmarks. We identified early studies that exemplified how ionizing radiation affects the hallmarks or how radiation was used experimentally to advance the understanding of key hallmarks. A literature search was performed to obtain relevant primary research, and topics were assigned to a particular hallmark to allow an organized, chronological account of the radiobiological advancements. The hallmarks are reviewed in an order that flows from cellular to microenvironmental effects.
Collapse
Affiliation(s)
- Mary-Keara Boss
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
| | - Robert Bristow
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University, Durham, North Carolina
- Address for correspondence: Duke University, Radiation Oncology, Room 201 MSRB, Research Drive, Durham, NC 27710;
| |
Collapse
|
19
|
Park SI, Lee C, Sadler WD, Koh AJ, Jones J, Seo JW, Soki FN, Cho SW, Daignault SD, McCauley LK. Parathyroid hormone-related protein drives a CD11b+Gr1+ cell-mediated positive feedback loop to support prostate cancer growth. Cancer Res 2013; 73:6574-83. [PMID: 24072746 DOI: 10.1158/0008-5472.can-12-4692] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the tumor microenvironment, CD11b(+)Gr1(+) bone marrow-derived cells are a predominant source of protumorigenic factors such as matrix metalloproteinases (MMP), but how distal tumors regulate these cells in the bone marrow is unclear. Here we addressed the hypothesis that the parathyroid hormone-related protein (PTHrP) potentiates CD11b(+)Gr1(+) cells in the bone marrow of prostate tumor hosts. In two xenograft models of prostate cancer, levels of tumor-derived PTHrP correlated with CD11b(+)Gr1(+) cell recruitment and microvessel density in the tumor tissue, with evidence for mediation of CD11b(+)Gr1(+) cell-derived MMP-9 but not tumor-derived VEGF-A. CD11b(+)Gr1(+) cells isolated from mice with PTHrP-overexpressing tumors exhibited relatively increased proangiogenic potential, suggesting that prostate tumor-derived PTHrP potentiates this activity of CD11b(+)Gr1(+) cells. Administration of neutralizing PTHrP monoclonal antibody reduced CD11b(+)Gr1(+) cells and MMP-9 in the tumors. Mechanistic investigations in vivo revealed that PTHrP elevated Y418 phosphorylation levels in Src family kinases in CD11b(+)Gr1(+) cells via osteoblast-derived interleukin-6 and VEGF-A, thereby upregulating MMP-9. Taken together, our results showed that prostate cancer-derived PTHrP acts in the bone marrow to potentiate CD11b(+)Gr1(+) cells, which are recruited to tumor tissue where they contribute to tumor angiogenesis and growth.
Collapse
Affiliation(s)
- Serk In Park
- Authors' Affiliations: Departments of Medicine and Cancer Biology; Center for Bone Biology; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry; Comprehensive Cancer Center Biostatistics Core; and Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov 2013; 12:526-42. [PMID: 23812271 DOI: 10.1038/nrd4003] [Citation(s) in RCA: 327] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Approximately 50% of all patients with cancer receive radiation therapy at some point during the course of their treatment, and the majority of these patients are treated with curative intent. Despite recent advances in the planning of radiation treatment and the delivery of image-guided radiation therapy, acute toxicity and potential long-term side effects often limit the ability to deliver a sufficient dose of radiation to control tumours locally. In the past two decades, a better understanding of the hallmarks of cancer and the discovery of specific signalling pathways by which cells respond to radiation have provided new opportunities to design molecularly targeted therapies to increase the therapeutic window of radiation therapy. Here, we review efforts to develop approaches that could improve outcomes with radiation therapy by increasing the probability of tumour cure or by decreasing normal tissue toxicity.
Collapse
|
21
|
Yang KL, Wang YS, Chang CC, Huang SC, Huang YC, Chi MS, Chi KH. Reciprocal complementation of the tumoricidal effects of radiation and natural killer cells. PLoS One 2013; 8:e61797. [PMID: 23634213 PMCID: PMC3636248 DOI: 10.1371/journal.pone.0061797] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/13/2013] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment is a key determinant for radio-responsiveness. Immune cells play an important role in shaping tumor microenvironments; however, there is limited understanding of how natural killer (NK) cells can enhance radiation effects. This study aimed to assess the mechanism of reciprocal complementation of radiation and NK cells on tumor killing. Various tumor cell lines were co-cultured with human primary NK cells or NK cell line (NK-92) for short periods and then exposed to irradiation. Cell proliferation, apoptosis and transwell assays were performed to assess apoptotic efficacy and cell viability. Western blot analysis and immunoprecipitation methods were used to determine XIAP (X-linked inhibitor of apoptosis protein) and Smac (second mitochondria-derived activator of caspase) expression and interaction in tumor cells. Co-culture did not induce apoptosis in tumor cells, but a time- and dose-dependent enhancing effect was found when co-cultured cells were irradiated. A key role for caspase activation via perforin/granzyme B (Grz B) after cell-cell contact was determined, as the primary radiation enhancing effect. The efficacy of NK cell killing was attenuated by upregulation of XIAP to bind caspase-3 in tumor cells to escape apoptosis. Knockdown of XIAP effectively potentiated NK cell-mediated apoptosis. Radiation induced Smac released from mitochondria and neutralized XIAP and therefore increased the NK killing. Our findings suggest NK cells in tumor microenvironment have direct radiosensitization effect through Grz B injection while radiation enhances NK cytotoxicity through triggering Smac release.
Collapse
Affiliation(s)
- Kai-Lin Yang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chao-Chun Chang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Su-Chen Huang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yi-Chun Huang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Institute of Radiation Science and School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
22
|
Park SI, Liao J, Berry JE, Li X, Koh AJ, Michalski ME, Eber MR, Soki FN, Sadler D, Sud S, Tisdelle S, Daignault SD, Nemeth JA, Snyder LA, Wronski TJ, Pienta KJ, McCauley LK. Cyclophosphamide creates a receptive microenvironment for prostate cancer skeletal metastasis. Cancer Res 2012; 72:2522-32. [PMID: 22589273 DOI: 10.1158/0008-5472.can-11-2928] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A number of cancers predominantly metastasize to bone, due to its complex microenvironment and multiple types of constitutive cells. Prostate cancer especially has been shown to localize preferentially to bones with higher marrow cellularity. Using an experimental prostate cancer metastasis model, we investigated the effects of cyclophosphamide, a bone marrow-suppressive chemotherapeutic drug, on the development and growth of metastatic tumors in bone. Priming the murine host with cyclophosphamide before intracardiac tumor cell inoculation was found to significantly promote tumor localization and subsequent growth in bone. Shortly after cyclophosphamide treatment, there was an abrupt expansion of myeloid lineage cells in the bone marrow and the peripheral blood, associated with increases in cytokines with myelogenic potential such as C-C chemokine ligand (CCL)2, interleukin (IL)-6, and VEGF-A. More importantly, neutralizing host-derived murine CCL2, but not IL-6, in the premetastatic murine host significantly reduced the prometastatic effects of cyclophosphamide. Together, our findings suggest that bone marrow perturbation by cytotoxic chemotherapy can contribute to bone metastasis via a transient increase in bone marrow myeloid cells and myelogenic cytokines. These changes can be reversed by inhibition of CCL2.
Collapse
Affiliation(s)
- Serk In Park
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Issues to be considered when studying cancer in vitro. Crit Rev Oncol Hematol 2012; 85:95-111. [PMID: 22823950 DOI: 10.1016/j.critrevonc.2012.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 05/31/2012] [Accepted: 06/27/2012] [Indexed: 01/17/2023] Open
Abstract
Various cancer treatment approaches have shown promising results when tested preclinically. The results of clinical trials, however, are often disappointing. While searching for the reasons responsible for their failures, the relevance of experimental and preclinical models has to be taken into account. Possible factors that should be considered, including cell modifications during in vitro cultivation, lack of both the relevant interactions and the structural context in vitro have been summarized in the present review.
Collapse
|
24
|
Chi KH, Wang YS, Kao SJ. Improving Radioresponse Through Modification of the Tumor Immunological Microenvironment. Cancer Biother Radiopharm 2012; 27:6-11. [DOI: 10.1089/cbr.2011.1048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Shang-Jyh Kao
- Division of Chest Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
25
|
Guo Y, Shu C, Zhou X, Jiang X, Li Q. High-dose preoperative fractionated radiotherapy does not affect the patency and healing of ePTFE vascular prosthesis after replacement of canine abdominal aorta and inferior vena cava. Eur J Vasc Endovasc Surg 2012; 43:460-5. [PMID: 22261487 DOI: 10.1016/j.ejvs.2011.12.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 12/20/2011] [Indexed: 12/01/2022]
Abstract
BACKGROUND Fractionated radiotherapy allows for the safe administration of larger doses without the development of immediate or late toxicity. The influence of preoperative fractionated radiotherapy on neointima formation for expanded polytetrafluoroethylene (ePTFE) graft has not been determined. METHODS Twenty mongrel dogs were randomly divided into radiotherapy group (a total dose of 35 Gy) or control group (no radiation). The infrarenal abdominal aorta and inferior vena cava were replaced by ePTFE grafts at 3 months after irradiation in the radiotherapy group. Grafts were explanted at 4 weeks after surgery. Histopathological techniques were undertaken to evaluate graft neointima formation. The control group was managed the same as the radiotherapy group except for not receiving irradiation. RESULTS Four grafts implanted into inferior vena cava in the irradiated group and three in the control group were found to be completely occluded. None of the grafts implanted into abdominal aorta were obstructed. In the case of the inferior vena cava graft, the thickness of the graft neointima did not differ significantly between the irradiated and control groups. However, for the abdominal aorta graft, the neointima thickness in the irradiated groups was significantly thinner than that in the control group. CONCLUSION Preoperative fractionated radiotherapy affects vascular healing via suppressing the development of neointima formation in the abdominal aorta graft.
Collapse
Affiliation(s)
- Y Guo
- Department of Vascular Surgery, The Second Xiang Ya Hospital, Central South University, 139 Renmin Road, Changsha, Hunan 410011, PR China
| | | | | | | | | |
Collapse
|
26
|
The confluence of stereotactic ablative radiotherapy and tumor immunology. Clin Dev Immunol 2011; 2011:439752. [PMID: 22162711 PMCID: PMC3227385 DOI: 10.1155/2011/439752] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 09/05/2011] [Indexed: 11/17/2022]
Abstract
Stereotactic radiation approaches are gaining more popularity for the treatment of intracranial as well as extracranial tumors in organs such as the liver and lung. Technology, rather than biology, is driving the rapid adoption of stereotactic body radiation therapy (SBRT), also known as stereotactic ablative radiotherapy (SABR), in the clinic due to advances in precise positioning and targeting. Dramatic improvements in tumor control have been demonstrated; however, our knowledge of normal tissue biology response mechanisms to large fraction sizes is lacking. Herein, we will discuss how SABR can induce cellular expression of MHC I, adhesion molecules, costimulatory molecules, heat shock proteins, inflammatory mediators, immunomodulatory cytokines, and death receptors to enhance antitumor immune responses.
Collapse
|
27
|
Shu C, Guo Y, Zhou X, Wan H, Yan J, Yuan L. Effect of postoperative fractionated radiotherapy on canine ePTFE graft neointima and anastomotic stoma healing: A preliminary experimental study. Asian J Surg 2011; 34:121-7. [DOI: 10.1016/j.asjsur.2011.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 07/03/2011] [Accepted: 07/18/2011] [Indexed: 10/16/2022] Open
|
28
|
Chou CH, Teng CM, Tzen KY, Chang YC, Chen JH, Cheng JCH. MMP-9 from sublethally irradiated tumor promotes Lewis lung carcinoma cell invasiveness and pulmonary metastasis. Oncogene 2011; 31:458-68. [DOI: 10.1038/onc.2011.240] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
29
|
Abstract
Radiotherapy is used to treat approximately 50% of all cancer patients, with varying success. The dose of ionizing radiation that can be given to the tumour is determined by the sensitivity of the surrounding normal tissues. Strategies to improve radiotherapy therefore aim to increase the effect on the tumour or to decrease the effects on normal tissues. These aims must be achieved without sensitizing the normal tissues in the first approach and without protecting the tumour in the second approach. Two factors have made such approaches feasible: namely, an improved understanding of the molecular response of cells and tissues to ionizing radiation and a new appreciation of the exploitable genetic alterations in tumours. These have led to the development of treatments combining pharmacological interventions with ionizing radiation that more specifically target either tumour or normal tissue, leading to improvements in efficacy.
Collapse
Affiliation(s)
- Adrian C Begg
- Division of Experimental Therapy, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands.
| | | | | |
Collapse
|
30
|
Sangaletti S, Tripodo C, Ratti C, Piconese S, Porcasi R, Salcedo R, Trinchieri G, Colombo MP, Chiodoni C. Oncogene-driven intrinsic inflammation induces leukocyte production of tumor necrosis factor that critically contributes to mammary carcinogenesis. Cancer Res 2010; 70:7764-75. [PMID: 20924115 PMCID: PMC7371347 DOI: 10.1158/0008-5472.can-10-0471] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Oncogene activation promotes an intrinsic inflammatory pathway that is crucial for cancer development. Here, we have investigated the actual effect of the inflammatory cytokine tumor necrosis factor (TNF) on the natural history of spontaneous mammary cancer in the HER2/neuT (NeuT) transgenic mouse model. Bone marrow transplantation from TNF knockout mice into NeuT recipients significantly impaired tumor growth, indicating that the source of TNF fostering tumor development was of bone marrow origin. We show that the absence of leukocyte-derived TNF disarranged the tumor vasculature, which lacked pericyte coverage and structural integrity, leading to diffuse vascular hemorrhage and stromal necrosis. In addition, tumor-associated Tie2-expressing monocytes were reduced and cytokine expression skewed from Th2 to Th1 type. Treatment of NeuT mice with anti-TNF antibody partially phenocopied the antitumor effect of TNF-deficient bone marrow cell transplantation, providing a strong preclinical background and rationale for the introduction of TNF antagonists in the treatment of human breast cancer, including basal-like samples for which consolidated targeted therapies do not exist.
Collapse
MESH Headings
- Animals
- Antibodies/therapeutic use
- Bone Marrow Transplantation
- Crosses, Genetic
- Female
- Humans
- Immunohistochemistry
- Inflammation/pathology
- Inflammation/physiopathology
- Leukocytes/physiology
- Male
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oncogenes
- Platelet Endothelial Cell Adhesion Molecule-1/analysis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/deficiency
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Tumor Necrosis Factor-alpha/deficiency
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Sabina Sangaletti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudio Tripodo
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Chiara Ratti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Silvia Piconese
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Rossana Porcasi
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Rosalba Salcedo
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Mario P. Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudia Chiodoni
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
31
|
Cubillos-Ruiz JR, Rutkowski M, Conejo-Garcia JR. Blocking ovarian cancer progression by targeting tumor microenvironmental leukocytes. Cell Cycle 2010; 9:260-8. [PMID: 20023378 DOI: 10.4161/cc.9.2.10430] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current therapies for metastatic ovarian carcinoma are based on surgical debulking followed by chemotherapy. After more than three decades implementing treatments that selectively target the tumor cell, the 5-year survival rate for metastatic ovarian cancer patients is still lower than 30%. Novel strategies are therefore urgently needed to complement classical treatments for this malignancy. Recently, leukocytes in the ovarian cancer microenvironment such as regulatory T cells and immature pro-angiogenic/tolerogenic myeloid cells have been demonstrated to play a fundamental role in tumor progression. This review focuses on our recent understanding of the potential of eliminating and/or modulating the phenotype of these leukocytes in vivo and in situ as a novel intervention to complement standard ovarian cancer treatments. The significant effects of targeting these crucial microenvironmental players on cancer vascularization, local tumor growth, distal metastatic spreading and spontaneous anti-tumor immune responses are discussed.
Collapse
|