1
|
Zhang Y, Cao S, Zeng F, Pan D, Cai L, Zhou Y, Wang H, Qin G, Zhang C, Chen W. Dihydroartemisinin enhances the radiosensitivity of breast cancer by targeting ferroptosis signaling pathway through hsa_circ_0001610. Eur J Pharmacol 2024; 983:176943. [PMID: 39182549 DOI: 10.1016/j.ejphar.2024.176943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVE This study aimed to elucidate how DHA enhances the radiosensitivity of BC and to explain its potential mechanisms of action. METHODS The circular structure of hsa_circ_0001610 was confirmed by Sanger sequencing, RNase R treatment, RT-PCR analysis using gDNA or cDNA. Cellular localization of hsa_circ_0001610 and microRNA-139-5p (miR-139-5p) was detected by fluorescence in situ hybridization. Cell counting kit-8 assay, wound healing and colony formation tests for assessing cell proliferation, while flow cytometry was utilized to estimate cell cycle progression and apoptosis. Reactive oxygen species and malondialdehyde experiments were conducted to validate ferroptosis of BC cells. The expression of ncRNAs and mRNAs was quantified via qRT-PCR, and protein expression was analyzed using Western blot. The effects of hsa_circ_0001610 and DHA on radiosensitivity of BC in vivo were studied by establishing BC mice model. RESULTS In vivo and in vitro experimental results indicate that DHA promotes ferroptosis of BC cells at least partly by inhibiting hsa_circ_0001610/miR-139-5p/SLC7A11 pathway, thereby enhancing the radiosensitivity of BC cells. CONCLUSIONS Our findings showed that DHA can induce ferroptosis of BC cells by down-regulation of hsa_circ_0001610, thus enhancing radiosensitivity, suggesting a promising therapeutic strategy for enhancing BC radiosensitivity that is worthy of further exploration.
Collapse
Affiliation(s)
- YiWen Zhang
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - ShuYi Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510000, China
| | - FengXia Zeng
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - DeRun Pan
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - LongMei Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - YingYing Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - HongMei Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - GengGeng Qin
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China.
| | - Chao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510000, China.
| | - WeiGuo Chen
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
2
|
Zumsteg ZS, Sheth S, Jabbour SK, Patel KR, Kimple RJ, Williams TM, Xu-Welliver M, Torres-Saavedra PA, Monjazeb AM, Mayadev J, Finkelstein SE, Buatti JM, Patel SP, Lin SH. Challenges and opportunities for early phase clinical trials of novel drug-radiotherapy combinations: recommendations from NRG Oncology, the American Society for Radiation Oncology (ASTRO), the American College of Radiology (ACR), the Sarah Cannon Research Institute, and the American College of Radiation Oncology (ACRO). Lancet Oncol 2024; 25:e489-e500. [PMID: 39362260 DOI: 10.1016/s1470-2045(24)00264-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 10/05/2024]
Abstract
NRG Oncology's Developmental Therapeutics and Radiation Therapy Subcommittee assembled an interdisciplinary group of investigators to address barriers to successful early phase clinical trials of novel combination therapies involving radiation. This Policy Review elucidates some of the many challenges associated with study design for early phase trials combining radiotherapy with novel systemic agents, which are distinct from drug-drug combination development and are often overlooked. We also advocate for potential solutions that could mitigate or eliminate some of these barriers, providing examples of specific clinical trial designs that could help facilitate efficient and effective evaluation of novel drug-radiotherapy combinations.
Collapse
Affiliation(s)
- Zachary S Zumsteg
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Siddharth Sheth
- Division of Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Krishnan R Patel
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Randall J Kimple
- Department of Human Oncology, Univeristy of Wisconsin, Madison, WI, USA
| | | | - Meng Xu-Welliver
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pedro A Torres-Saavedra
- Division of Cancer Treatment and Diagnosis, Biometric Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California, San Diego, CA, USA
| | - Jyoti Mayadev
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, CA, USA
| | - Steven E Finkelstein
- The US Oncology Network, Florida Cancer Affiliates, Panama City, FL, USA; Sarah Cannon Research Institute, Nashville, TN, USA; Associated Medical Professional of NY, US Urology Partners, Syracuse, NY, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Sandip P Patel
- Division of Medical Oncology, University of California, San Diego, CA, USA
| | - Steven H Lin
- Department of Thoracic Radiation Oncology, Division of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Gilbert A, Samuel R, Cagney D, Sebag-Montefiore D, Brown J, Brown SR. The use of master protocols for efficient trial design to evaluate radiotherapy interventions: a systematic review. J Natl Cancer Inst 2024; 116:1220-1229. [PMID: 38720568 PMCID: PMC11308198 DOI: 10.1093/jnci/djae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/05/2024] [Accepted: 04/07/2024] [Indexed: 08/09/2024] Open
Abstract
The aim of this review was to highlight why the use of master protocols trial design is particularly useful for radiotherapy intervention trials where complex setup pathways (including quality assurance, user training, and integrating multiple modalities of treatment) may hinder clinical advances. We carried out a systematic review according to Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines, reviewing the findings using a landscape analysis. Results were summarized descriptively, reporting on trial characteristics highlighting the benefits, limitations, and challenges of developing and implementing radiotherapy master protocols, with three case studies selected to explore these issues in more detail. Twelve studies were suitable for inclusion (4 platform trials, 3 umbrella trials, and 5 basket trials), evaluating a mix of solid tumor sites in both curative and palliative settings. The interventions were categorized into 1) novel agent and radiotherapy combinations; 2) radiotherapy dose personalization; and 3) device evaluation, with a case study provided for each intervention. Benefits of master protocol trials for radiotherapy intervention include protocol efficiency for implementation of novel radiotherapy techniques; accelerating the evaluation of novel agent drug and radiotherapy combinations; and more efficient translational research opportunities, leading to cost savings and research efficiency to improve patient outcomes. Master protocols offer an innovative platform under which multiple clinical questions can be addressed within a single trial. Due to the complexity of radiotherapy trial setup, cost and research efficiency savings may be more apparent than in systemic treatment trials. Use of this research approach may be the change needed to push forward oncological innovation within radiation oncology.
Collapse
Affiliation(s)
- Alexandra Gilbert
- Leeds Institute for Medical Research, University of Leeds, St James’s University Hospital, Leeds, UK
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Robert Samuel
- Leeds Institute for Medical Research, University of Leeds, St James’s University Hospital, Leeds, UK
| | - Daniel Cagney
- Radiation Oncology, Mater Private Hospital, Dublin, Ireland
- Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - David Sebag-Montefiore
- Leeds Institute for Medical Research, University of Leeds, St James’s University Hospital, Leeds, UK
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Julia Brown
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Sarah R Brown
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| |
Collapse
|
4
|
Sharma P, Maurya DK. Wharton's jelly mesenchymal stem cells: Future regenerative medicine for clinical applications in mitigation of radiation injury. World J Stem Cells 2024; 16:742-759. [PMID: 39086560 PMCID: PMC11287430 DOI: 10.4252/wjsc.v16.i7.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/28/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Wharton's jelly mesenchymal stem cells (WJ-MSCs) are gaining significant attention in regenerative medicine for their potential to treat degenerative diseases and mitigate radiation injuries. WJ-MSCs are more naïve and have a better safety profile, making them suitable for both autologous and allogeneic transplantations. This review highlights the regenerative potential of WJ-MSCs and their clinical applications in mitigating various types of radiation injuries. In this review, we will also describe why WJ-MSCs will become one of the most probable stem cells for future regenerative medicine along with a balanced view on their strengths and weaknesses. Finally, the most updated literature related to both preclinical and clinical usage of WJ-MSCs for their potential application in the regeneration of tissues and organs will also be compiled.
Collapse
Affiliation(s)
- Prashasti Sharma
- Life Sciences, Homi Bhabha National Institute, Mumbai 400094, Maharashtra, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
| | - Dharmendra Kumar Maurya
- Life Sciences, Homi Bhabha National Institute, Mumbai 400094, Maharashtra, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India.
| |
Collapse
|
5
|
Mahadevan J, Appudurai R, Sothipragasam S, Kumar R, Rajasooriyar C. "Current", "heated rods", and "hot vapour": why patients refuse radiotherapy as a treatment modality for cancer in northern Sri Lanka. Support Care Cancer 2024; 32:361. [PMID: 38753165 DOI: 10.1007/s00520-024-08561-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 05/09/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE Significant proportions of patients either refuse or discontinue radiotherapy, even in the curative setting, leading to poor clinical outcomes. This study explores patient perceptions that underlie decisions to refuse/discontinue radiotherapy at a cancer care facility in northern Sri Lanka. METHODS An exploratory descriptive qualitative study was carried out among 14 purposively selected patients with cancer who refused/discontinued radiotherapy. In-depth semi-structured interviews were transcribed in Tamil, translated into English, coded, and thematically analyzed. RESULTS All participants referred to radiotherapy as "current" with several understanding the procedure to involve electricity, heat, or hot vapour. Many pointed to gaps in information provided by healthcare providers, who were perceived to focus on side effects without explaining the procedure. In the absence of these crucial details, patients relied on family members and acquaintances for information, often based on second or third-hand accounts of experiences with radiotherapy. Many felt pressured by family to refuse radiation, feared radiation, or felt ashamed to ask questions, while for others COVID-19 was an impediment. All but three participants regretted their decision, claiming they would recommend radiation to patients with cancer, especially when it is offered with curative intent. CONCLUSION Patients with cancer who refused/discontinued radiation therapy have significant information needs. While human resource deficits need to be addressed in low-resource settings like northern Sri Lanka, providing better supportive cancer care could improve clinical outcomes and save healthcare resources that would otherwise be wasted on patient preparation for radiotherapy.
Collapse
Affiliation(s)
| | - Ramalingam Appudurai
- Department of Clinical Oncology, Hospital Road, Teaching Hospital Jaffna, Jaffna, 40 000, Sri Lanka
- Tellipalai Trail Cancer Hospital, Tellipalai, Sri Lanka
| | | | - Ramya Kumar
- Department of Community and Family Medicine, University of Jaffna, Jaffna, Sri Lanka
| | - Chrishanthi Rajasooriyar
- Department of Clinical Oncology, Hospital Road, Teaching Hospital Jaffna, Jaffna, 40 000, Sri Lanka.
- Tellipalai Trail Cancer Hospital, Tellipalai, Sri Lanka.
| |
Collapse
|
6
|
Zarakowska E, Guz J, Mijewski P, Wasilow A, Wozniak J, Roszkowski K, Foksinski M, Gackowski D, Olinski R. Intracellular ascorbate is a safe-guard and/or reservoir for plasma vitamin C in prostate cancer patients undergoing radiotherapy. Free Radic Biol Med 2024; 210:230-236. [PMID: 38036068 DOI: 10.1016/j.freeradbiomed.2023.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/03/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
Prostate cancer (PC) represents one of the most common cancer types worldwide and many patients suffering from this kind of cancer are treated with radiotherapy (RTH). Ionizing irradiation is closely associated with reactive oxygen species (ROS) production and oxidative stress. Over the years the role of vitamin C (VC) in cancer prevention has been highlighted as it may be mediated by its ability to neutralize pro-carcinogenic ROS. However, the debate concerning the presence of VC in blood and its beneficial effect on the survival of cancer patients is inconsistent and controversial. To our best knowledge until recently there have been no studies concerning such a role of intracellular VC (iVC). In the present study, blood and intracellular concentrations of vitamin C were analyzed along with the level of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), as an established marker of the stress condition, in leukocytes of PC patients during the course of radiotherapy. The level of intracellular vitamin C significantly decreased in PC patients in comparison with the healthy group, while there were no differences in blood VC. It was observed that a sub-group of the PC patients reacted to RTH decreasing VC in leukocytes (group A), while the other sub-group acted the other way round, significantly increasing its level (group B). Under stressful conditions (RTH) leukocytes react in two different ways. Both ways are in good agreement with two well recognized functions, proposed for iVC; it may serve as a save factor, to protect the cellular DNA, increasing its concentration inside the cell (group B), and as a reservoir decreasing the VC level inside leukocytes and releasing VC into the plasma to rescue its physiological level (group A). It was also demonstrated that there was a relationship between the level of 8-oxodG in leukocytes' DNA and the markers of RTH toxicity.
Collapse
Affiliation(s)
- Ewelina Zarakowska
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Jolanta Guz
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Pawel Mijewski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Aleksandra Wasilow
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Jakub Wozniak
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Krzysztof Roszkowski
- Department of Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-821 Bydgoszcz, Poland.
| | - Marek Foksinski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Daniel Gackowski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| | - Ryszard Olinski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland.
| |
Collapse
|
7
|
Ahmedah HT, Basheer HA, Almazari I, Amawi KF. Introduction to Nutrition and Cancer. Cancer Treat Res 2024; 191:1-32. [PMID: 39133402 DOI: 10.1007/978-3-031-55622-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
By the beginning of the year 2021, the estimated number of new cancer cases worldwide was about 19.3 million and there were 10.0 million cancer-related deaths. Cancer is one of the deadliest diseases worldwide that can be attributed to genetic and environmental factors, including nutrition. The good nutrition concept focuses on the dietary requirements to sustain life. There is a substantial amount of evidence suggesting that a healthy diet can modulate cancer risk, particularly the risk of colorectal and breast cancers. Many studies have evaluated the correlation between our diet and the risk of cancer development, prevention, and treatment. The effect of diet on cancer development is likely to happen through intertwining mechanisms including inflammation and immune responses. For instance, a greater intake of red and processed meat along with low consumption of fruits and vegetables has been associated with increased levels of inflammatory biomarkers that are implicated in cancer development. On the other hand, the consumption of phytosterols, vitamins, and minerals, which exert antioxidant and anti-inflammatory roles have been linked to lower cancer risk, or even its occurrence prevention. In this book, we aim to summarize the current knowledge on the role of nutrition in cancer to provide the best scientific advice in this regard.
Collapse
Affiliation(s)
- Hanadi Talal Ahmedah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Rabigh, 25732, Saudi Arabia.
| | | | - Inas Almazari
- Department of Clinical Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| | - Kawther Faisal Amawi
- Department of Medical Laboratory Science, Faculty of Allied Medical Science, Zarqa University, PO Box 132222, Zarqa, 13132, Jordan
| |
Collapse
|
8
|
Katolkar UN, Surana SJ. Exploring the Potential Role of Phytopharmaceuticals in Alleviating Toxicities of Chemotherapeutic Agents. Curr Protein Pept Sci 2024; 25:753-779. [PMID: 38919003 DOI: 10.2174/0113892037307940240606075208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Chemotherapy is the mainstay of cancer treatment, bringing patients optimism about recurrence and survival. However, the clinical effectiveness of chemotherapeutic drugs is frequently jeopardized by their intrinsic toxicity, resulting in side effects affecting the quality of life of cancer patients. This analysis explores the ethnopharmacological impact of phytopharmaceuticals, highlighting their traditional use in many cultures. The present study, which takes its cues from indigenous knowledge, aims to close the knowledge gap between traditional medicine and modern medicine in reducing the toxicities of chemotherapy treatments. AIM The present in-depth study aims to highlight the current research and upcoming developments in phytopharmaceuticals for reducing the toxicity of chemotherapeutic drugs. Further, we address the mechanisms through which phytopharmaceuticals may reduce chemotherapy-induced side effects that include nausea, vomiting, myelosuppression, nephropathy, neuropathy, and cardiotoxicity using data from a variety of preclinical and clinical investigations. MATERIALS AND METHODS The literature search was carried out by employing search engines such as PubMed and Google Scholar with keywords such as cancer, chemotherapy, CNS toxicity, hematopoietic toxicity, renal toxicity, GI toxicity, CNS toxicity, and phytopharmaceuticals. RESULTS Bioactive chemicals found in plants, such as fruits, vegetables, herbs, and spices, are being studied for their capacity to improve the safety and acceptability of chemotherapy regimens. The current review also dives into the investigation of phytopharmaceuticals as adjuvant medicines in cancer treatment, which is a viable path for addressing the pressing need to lessen chemotherapy-induced toxicities. CONCLUSION The present review revealed that the potential of phytopharmaceuticals in alleviating chemotherapeutic drug toxicities would pave the way for better cancer treatment and patient outcomes, harmonizing with the larger trend towards personalized and holistic approaches to chemotherapy.
Collapse
Affiliation(s)
- Ujwal N Katolkar
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| | - Sanjay J Surana
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| |
Collapse
|
9
|
Markov AG, Livanova AA, Fedorova AA, Kravtsova VV, Krivoi II. Chronic Ouabain Targets Pore-Forming Claudin-2 and Ameliorates Radiation-Induced Damage to the Rat Intestinal Tissue Barrier. Int J Mol Sci 2023; 25:278. [PMID: 38203449 PMCID: PMC10778734 DOI: 10.3390/ijms25010278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Ionizing radiation (IR) causes disturbances in the functions of the gastrointestinal tract. Given the therapeutic potential of ouabain, a specific ligand of the Na,K-ATPase, we tested its ability to protect against IR-induced disturbances in the barrier and transport properties of the jejunum and colon of rats. Male Wistar rats were subjected to 6-day intraperitoneal injections of vehicle or ouabain (1 µg/kg/day). On the fourth day of injections, rats were exposed to total-body X-ray irradiation (10 Gy) or a sham irradiation. Isolated tissues were examined 72 h post-irradiation. Electrophysiological characteristics and paracellular permeability for sodium fluorescein were measured in an Ussing chamber. Histological analysis and Western blotting were also performed. In the jejunum tissue, ouabain exposure did not prevent disturbances in transepithelial resistance, paracellular permeability, histological characteristics, as well as changes in the expression of claudin-1, -3, -4, tricellulin, and caspase-3 induced by IR. However, ouabain prevented overexpression of occludin and the pore-forming claudin-2. In the colon tissue, ouabain prevented electrophysiological disturbances and claudin-2 overexpression. These observations may reveal a mechanism by which circulating ouabain maintains tight junction integrity under IR-induced intestinal dysfunction.
Collapse
Affiliation(s)
- Alexander G. Markov
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.A.L.); (A.A.F.); (V.V.K.); (I.I.K.)
| | | | | | | | | |
Collapse
|
10
|
Mela A, Rdzanek E, Jaroszyński J, Furtak-Niczyporuk M, Jabłoński M, Niewada M. Reimbursement decision-making system in Poland systematically compared to other countries. Front Pharmacol 2023; 14:1153680. [PMID: 37900165 PMCID: PMC10611478 DOI: 10.3389/fphar.2023.1153680] [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: 01/29/2023] [Accepted: 08/24/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: Our objective was to analyze and compare systematically and structurally reimbursement systems in Poland and other countries. Methods: The systems were selected based on recommendations issued by the Polish Agency for Health Technology Assessment and Tariffication (AHTAPol), which explicitly referred to other countries and agencies). Consequently, apart from Poland, the countries included in the analysis were England, Scotland, Wales, Ireland, France, Netherlands, Germany, Norway, Sweden, Canada, Australia and New Zealand. Relevant information and data were collected through a systematic search of PubMed (Medline), Embase and The Cochrane Library as well as competent authority websites and grey literature sources. Results and discussion: In most of the countries, the submission of a reimbursement application is initiated by a pharmaceutical company, and only a few countries allow it before a product is approved for marketing. All of the agencies analyzed are independent and some have regulatory function of reimbursement decision making body. A key criterion differentiating the various agencies in terms of HTA is the cost-effectiveness threshold. Most of the countries have specific mechanisms to improve access to expensive specialty drugs, including cancer drugs and those used for rare diseases. Reimbursement systems often lack consistency in appreciating the same stages, leading to heterogeneous decision-making processes. The analysis of recommendations issued in different countries for the same medicinal product will allow a better understanding of the relations between the reimbursement system, HTA assessment, stakeholders involvement and decision on reimbursement of innovative drugs.
Collapse
Affiliation(s)
- Aneta Mela
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Elżbieta Rdzanek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Janusz Jaroszyński
- Department of Administrative Proceedings, Faculty of Law and Administration, Marie Curie-Sklodowska University, Lublin, Poland
| | | | - Mirosław Jabłoński
- Department of Orthopeadics and Rehabilitation, Medical University of Lublin, Lublin, Poland
| | - Maciej Niewada
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
11
|
Torres-Roca JF, Grass GD, Scott JG, Eschrich SA. Towards Data Driven RT Prescription: Integrating Genomics into RT Clinical Practice. Semin Radiat Oncol 2023; 33:221-231. [PMID: 37331777 DOI: 10.1016/j.semradonc.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The genomic era has significantly changed the practice of clinical oncology. The use of genomic-based molecular diagnostics including prognostic genomic signatures and new-generation sequencing has become routine for clinical decisions regarding cytotoxic chemotherapy, targeted agents and immunotherapy. In contrast, clinical decisions regarding radiation therapy (RT) remain uninformed about the genomic heterogeneity of tumors. In this review, we discuss the clinical opportunity to utilize genomics to optimize RT dose. Although from the technical perspective, RT has been moving towards a data-driven approach, RT prescription dose is still based on a one-size-fits all approach, with most RT dose based on cancer diagnosis and stage. This approach is in direct conflict with the realization that tumors are biologically heterogeneous, and that cancer is not a single disease. Here, we discuss how genomics can be integrated into RT prescription dose, the clinical potential for this approach and how genomic-optimization of RT dose could lead to new understanding of the clinical benefit of RT.
Collapse
Affiliation(s)
- Javier F Torres-Roca
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL; Department of Bioinformatics and Biostatistics, Moffitt Cancer Center, Tampa, FL; Department of Oncologic Sciences, University of South Florida College of Medicine, Tampa, FL.
| | - G Daniel Grass
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL; Department of Oncologic Sciences, University of South Florida College of Medicine, Tampa, FL
| | - Jacob G Scott
- Translational Hematology and Oncology Research, Radiation Oncology Department, Cleveland Clinic, Cleveland, OH
| | - Steven A Eschrich
- Department of Bioinformatics and Biostatistics, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
12
|
Arif M, Nawaz AF, Ullah khan S, Mueen H, Rashid F, Hemeg HA, Rauf A. Nanotechnology-based radiation therapy to cure cancer and the challenges in its clinical applications. Heliyon 2023; 9:e17252. [PMID: 37389057 PMCID: PMC10300336 DOI: 10.1016/j.heliyon.2023.e17252] [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: 03/29/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023] Open
Abstract
Radiation therapy against cancer frequently fails to attain the desired outcomes because of several restricting aspects. Radiation therapy is not a targeted antitumor treatment, and it poses serious threats to normal tissues as well. In many cases, some inherent features of tumors make them resistant to radiation therapy. Several nanoparticles have shown the capacity to upgrade the viability of radiation treatment because they can directly interact with ionizing radiation to increase cellular radiation sensitivity. Several types of nanomaterials have been investigated as radio-sensitizers, to improve the efficacy of radiotherapy and overcome radio-resistance including, metal-based nanoparticles, quantum dots, silica-based nanoparticles, polymeric nanoparticles, etc. Despite all this research and development, certain challenges associated with the exploitation of nanoparticles to enhance and improve radiation therapy for cancer treatment are encountered. Potential applications of nanoparticles as radiosensitizers is hindered by the difficulties associated with ensuring their production at a large scale with improved characterizations and because of certain biological challenges. By overcoming the shortcomings of nanoparticles like working on the pharmacokinetics, and physical and chemical characterization, the therapy can be improved. It is expected that in the future more knowledge will be available regarding nanoparticles and their clinical efficacy, leading to the successful development of nanotechnology-based radiation therapies for a variety of cancers. This review highlights the limitations of conventional radiotherapy in cancer treatment and explores the potential of nanotechnology, specifically the use of nanomaterials, to overcome these challenges. It discusses the concept of using nanomaterials to enhance the effectiveness of radiation therapy and provides an overview of different types of nanomaterials and their beneficial properties. The review emphasizes the need to address the obstacles and limitations associated with the application of nanotechnology in cancer radiation therapy for successful clinical translation.
Collapse
Affiliation(s)
- Muhammad Arif
- Department of Plant Biology and Ecology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Ayesha Fazal Nawaz
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Centre (NARC), Islamabad, Pakistan
| | - Shahid Ullah khan
- Department of Biochemistry, Women Medical and Dental College, Khyber Medical University KPK, Pakistan
| | - Hasnat Mueen
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Fizza Rashid
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Al-Medinah Al-Monawara Postcode, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
13
|
Chakka R, Vadaguru Dakshinamurthy R, Rawal P, Belladamadagu Appajappa S, Pramanik S. Gallic acid a flavonoid isolated from Euphorbia hirta antagonizes gamma radiation induced radiotoxicity in lymphocytes in vitro. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 20:146-152. [PMID: 36398419 DOI: 10.1515/jcim-2022-0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/21/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES The current study was executed to isolate and evaluate gallic acid from Euphorbia hirta for in vitro radioprotective potentials against gamma irradiation caused radiotoxicity in human lymphocytes. METHODS The defatted E. hirta plant material was treated to methanol extraction using the soxhlet device. Bioflavonoids were isolated from the E. hirta methanol extract using column chromatography. In human cells exhibited to gamma radiation, separated flavonoid gallic acid was examined for in vitro radioprotective potentials using the micronucleus test, DNA fragmentation assay, superoxide free radical scavenging method, and apoptic assay. RESULTS The frequency of micronuclei was considerably declined when cells were preprocessed with gallic acid (25 g/mL) before being exhibited to 2 Gy gamma radiation, as determined by the cytokinesis blocked micronucleus test. Similarly, pre-gamma radiation treatment of human cells with gallic acid led in markedly less DNA injury, as assessed by comet metrics like olive tail moment and percent tail DNA. Gallic acid (25 g/mL) given to lymphocytes prior to gamma irradiation considerably decreased the percentage of apoptotic bodies. Gallic acid also considerably lowered the reactive oxygen species concentrations elicited by gamma radiation. CONCLUSIONS Our findings showed that gallic acid protects lymphocytes isolated from human blood from gamma radiation-induced DNA destruction and anti-apoptotic activity, which could be because of inhibition of free radicals formed by gamma radiation as well as the decline of gamma radiation-induced oxidative stress.
Collapse
Affiliation(s)
- Ramesh Chakka
- Department of Pharmacology, East West College of Pharmacy, Bangalore, India
| | | | - Pinkey Rawal
- Department of Pharmaceutical Chemistry, East West College of Pharmacy, Bangalore, India
| | | | - Soma Pramanik
- Department of Pharmaceutical Chemistry, East West College of Pharmacy, Bangalore, India
| |
Collapse
|
14
|
Predicting tumour radiosensitivity to deliver precision radiotherapy. Nat Rev Clin Oncol 2023; 20:83-98. [PMID: 36477705 DOI: 10.1038/s41571-022-00709-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Abstract
Owing to advances in radiotherapy, the physical properties of radiation can be optimized to enable individualized treatment; however, optimization is rarely based on biological properties and, therefore, treatments are generally planned with the assumption that all tumours respond similarly to radiation. Radiation affects multiple cellular pathways, including DNA damage, hypoxia, proliferation, stem cell phenotype and immune response. In this Review, we summarize the effect of these pathways on tumour responses to radiotherapy and the current state of research on genomic classifiers designed to exploit these variations to inform treatment decisions. We also discuss whether advances in genomics have generated evidence that could be practice changing and whether advances in genomics are now ready to be used to guide the delivery of radiotherapy alone or in combination.
Collapse
|
15
|
Kilic SS, Mayo ZS, Weleff J, Parker S, Strzalka C, Phelan MP, Suh JH, Campbell SR, Shah CS. Cancer Diagnoses and Use of Radiation Therapy Among Persons Experiencing Homelessness. Int J Radiat Oncol Biol Phys 2023; 116:79-86. [PMID: 36731679 DOI: 10.1016/j.ijrobp.2023.01.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
PURPOSE Persons experiencing homelessness (PEH) have low rates of cancer screening and worse cancer mortality compared with persons not experiencing homelessness. Data regarding cancer diagnosis and treatment in PEH are limited. We investigated cancer prevalence and use of radiation therapy (RT) in PEH. METHODS AND MATERIALS Patients presenting between January 1, 2014, and September 27, 2021, at a large metropolitan hospital system were assessed for homelessness via intake screening or chart search. PEH data were cross-referenced with the institution's cancer database to identify PEH with cancer diagnoses. Demographic, clinical, and treatment variables were abstracted. RESULTS Of a total of 9654 (9250 evaluable) PEH with a median age of 42 years, 81 patients (0.88%) had at least 1 cancer diagnosis and 5 had multiple diagnoses, for a total of 87 PEH with at least 1 cancer diagnosis. The median age at diagnosis was 60 years. In total, 43% were female and 51% were Black, and 43% presented with advanced or metastatic disease. Lung (17%), prostate (15%), leukemia/lymphoma (13%), and head/neck (9%) were the most common diagnoses. In total, 17% of patients underwent surgery alone, 13% received chemotherapy alone, 14% received RT alone, and 6% received hormone therapy alone. A total of 8% of patients underwent no treatment, and 43% underwent multimodality therapy. In total, 58% of treated patients never achieved disease-free status. Of the 31 patients who received RT, 87% received external beam RT. Most patients (70%) received hypofractionated regimens. For patients who had multifraction treatment, the treatment completion rate was 85%, significantly lower than the departmental completion rate of 98% (P < .00001). CONCLUSIONS In a large cohort of PEH in a metropolitan setting, cancer diagnoses were uncommon and were frequently in advanced stages. Most patients underwent single-modality treatment or no treatment at all. Despite the use of hypofractionation, the RT completion rate was low, likely reflecting complex barriers to care. Further interventions to optimize cancer diagnosis and treatment in PEH are urgently needed.
Collapse
Affiliation(s)
- Sarah S Kilic
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio.
| | - Zachary S Mayo
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jeremy Weleff
- Department of Psychiatry and Psychology, Center for Behavioral Health, Neurological Institute, Cleveland Clinic, Cleveland, Ohio; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Sean Parker
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | - John H Suh
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Shauna R Campbell
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Chirag S Shah
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
16
|
Weng J, Zhang Y, Liang W, Xie Y, Wang K, Xu Q, Ding Y, Li Y. Downregulation of CEMIP enhances radiosensitivity by promoting DNA damage and apoptosis in colorectal cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2023; 40:73. [PMID: 36607478 DOI: 10.1007/s12032-022-01940-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023]
Abstract
Colorectal cancer (CRC) is the third leading malignancy worldwide in both new cases and deaths. Neoadjuvant radiotherapy is the standard preoperative regimens for locally advanced patients. However, approximately 50% of patients develop recurrence and metastasis after radiotherapy, which is largely due to the radiation resistance properties of the tumor, and the internal mechanism has not been elucidated. Here we found that CEMIP expression is up-regulated in a variety of tumor types, particularly in CRC. Public databases and clinical samples revealed that CEMIP expression is significantly higher in tumor tissues than in adjacent normal tissues in patients with locally advanced CRC who received neoadjuvant chemoradiotherapy, and it is closely related to the poor prognosis. Functional characterization uncovered that downregulation of CEMIP expression can enhance the radiosensitivity of CRC cells, which is confirmed to be achieved by promoting DNA damage and apoptosis. In vivo studies further verified that CEMIP knockdown can significantly improve the radiosensitivity of subcutaneously implanted colorectal tumors in mice, suggesting that CEMIP may be a radiation-resistant gene in CRC. Mechanistically, EGFR/PI3K/Akt signaling pathway is hypothesized to play a key role in CEMIP mediating radiation resistance. These results provide a potential new strategy targeting CEMIP gene for the comprehensive treatment of locally advanced CRC patients.
Collapse
Affiliation(s)
- Jiawen Weng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuqin Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weijie Liang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuwen Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kai Wang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qian Xu
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yiyi Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
17
|
Liu H, Miyamoto N, Nguyen MT, Shirato H, Yonezawa T. Injectable Fiducial Marker for Image-Guided Radiation Therapy Based on Gold Nanoparticles and a Body Temperature-Activated Gel-Forming System. ACS APPLIED BIO MATERIALS 2022; 5:4838-4848. [PMID: 36074396 DOI: 10.1021/acsabm.2c00566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Injectable fiducial markers are crucial in image-guided radiation therapy (IGRT) due to their minimally invasive operations and improved patient compliance. This study presents the development of a ready-to-use injectable fiducial marker utilizing alginate stabilized-gold nanoparticles (alg-Au NPs) and a body temperature-activated in situ gel-forming system. Gram-scale alg-Au NPs were prepared in an hour by a green microwave-induced plasma-in-liquid process (MWPLP). Sodium alginate was introduced in this process to avoid aggregation between Au NPs, which ensured their stability and injectability. The gelation behavior of alginate with divalent cations and a temperature-dependent release of calcium source (glucono-delta-lactone (GDL) and CaCO3) served as the foundation of the body temperature-activated in situ gel-forming system. The injectable fiducial marker GDL/CaCO3/alg-Au NPs could maintain a liquid state at a low temperature for a higher injectability. After injection, on the other hand, Ca2+ would be released due to the body temperature-activated hydrolysis of GDL and the subsequent reaction with CaCO3, which would initiate the gelation of alginate. The injectable fiducial marker can be therefore delivered via injection and form gel at target site to avoid marker movement or Au NPs leakage after injection. Rheological measurements demonstrate the stability and gelation behavior of GDL/CaCO3/alg-Au NPs at different temperatures. Furthermore, the injectability and imaging ability of GDL/CaCO3/alg-Au NPs were also examined. In summary, ready-to-use injectable fiducial marker GDL/CaCO3/alg-Au NPs were developed via a green and facile method for IGRT.
Collapse
Affiliation(s)
- Haoran Liu
- Division of Materials Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Naoki Miyamoto
- Division of Quantum Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
- Department of Medical Physics, Hokkaido University Hospital, Kita 14 Nishi 5, Kita-ku, Sapporo, Hokkaido 060-8648, Japan
| | - Mai Thanh Nguyen
- Division of Materials Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Tetsu Yonezawa
- Division of Materials Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| |
Collapse
|
18
|
Roychoudhury S, Das A, Panner Selvam MK, Chakraborty S, Slama P, Sikka SC, Kesari KK. Recent Publication Trends in Radiotherapy and Male Infertility over Two Decades: A Scientometric Analysis. Front Cell Dev Biol 2022; 10:877079. [PMID: 35646894 PMCID: PMC9133602 DOI: 10.3389/fcell.2022.877079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/06/2022] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy, a popular cancer management procedure, negatively impacts reproductive health particularly by reducing the fertility potential. The purpose of this study was to analyze the research trend in radiotherapy associated with male infertility over the past 20 years (2000-May 2021). SCOPUS database was used to retrieve relevant scientometric data (publication per year, affiliation, journals, countries, type of document and area of research) for different subgenres of radiotherapy and male infertility. A total of 275 articles were published related to radiotherapy and male infertility, with the United States being the most dominant country in research output in this field. Radiotherapy and male infertility research have shown positive growth over the last two decades. In-depth analysis revealed that publications (n) related to radiotherapy and male infertility research mainly focused its impact on semen parameters (n = 155) and fertility preservation techniques (n = 169). Our scientometric results highlight a limited research focus on the field of radiotherapy and its impact on male reproductive hormones. Furthermore, a significant lack of research was noticed in the area of omics and male reproductive organs linked to radiotherapy. Substantial research is warranted to further decipher the effect of radiotherapy, at molecular level, leading to male infertility.
Collapse
Affiliation(s)
- Shubhadeep Roychoudhury
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
- *Correspondence: Shubhadeep Roychoudhury, ; Manesh Kumar Panner Selvam,
| | - Anandan Das
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | - Manesh Kumar Panner Selvam
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, United States
- *Correspondence: Shubhadeep Roychoudhury, ; Manesh Kumar Panner Selvam,
| | | | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of Agrisciences, Mendel University in Brno, Brno, Czechia
| | - Suresh C. Sikka
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, United States
| | | |
Collapse
|
19
|
Ansari J, Farrag A, Ali A, Abdelgelil M, Murshid E, Alhamad A, Ali M, Ansari H, Hussain S, Glaholm J. Concurrent use of nivolumab and radiotherapy for patients with metastatic non-small cell lung cancer and renal cell carcinoma with oligometastatic disease progression on nivolumab. Mol Clin Oncol 2021; 15:214. [PMID: 34476098 PMCID: PMC8408674 DOI: 10.3892/mco.2021.2376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 07/09/2021] [Indexed: 11/22/2022] Open
Abstract
Checkpoint inhibitors (CPIs), such as nivolumab, have transformed the treatment paradigm for patients with metastatic non-small cell lung cancer (mNSCLC) and metastatic renal cell carcinoma (mRCC). The combination of CPIs and radiotherapy (RT) constitutes a multimodal treatment approach that may work synergistically and facilitate augmented systemic responses. The aim of the present retrospective study was to assess the efficacy and safety of continuation of nivolumab treatment with the addition of RT in patients with mNSCLC and mRCC who develop oligometastatic disease progression on single-agent nivolumab. All patients with mNSCLC and mRCC who received nivolumab at the Department of Oncology, Prince Sultan Military Medical City (Riyadh, Saudi Arabia) between November 2016 and April 2018 were identified. The records of patients who developed oligometastatic disease progression during nivolumab treatment and were subsequently treated with RT, with nivolumab continued beyond disease progression, were retrospectively reviewed. Details of RT, clinical outcomes and toxicity data were collected. Of the 96 patients who received nivolumab, 22 received multiple courses of RT. A total of 39 sites were irradiated: Bone (n=15), lung (n=9), brain (n=8), adrenal gland (n=2), renal bed (n=2), skin (n=1), ethmoid sinus (n=1) and scalp (n=1). Partial response and complete response were noted at 25 (64%) and 3 (8%) sites, respectively. Stable disease was noted at 6 sites (15%) and disease progression was noted at 5 sites (13%). The median time on nivolumab from the date of the first fraction of RT was 4.5 months (range, 1.5-29 months) for patients with mNSCLC and 5 months (range, 1-38.5 months) for patients with mRCC. No patients developed grade 3-4 toxicities. Grade 2 pneumonitis was noted in 3 patients receiving lung RT. The addition of RT appeared to initiate a response and prolong the duration of nivolumab treatment. Therefore, the combination of nivolumab and RT was found to be well tolerated, with response rates exceeding those in published studies of nivolumab monotherapy.
Collapse
Affiliation(s)
- Jawaher Ansari
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia.,Department of Oncology, Tawam Hospital, Al Ain 15258, United Arab Emirates
| | - Ashraf Farrag
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia.,Clinical Oncology Department, Assiut University Hospital, Assiut 71515, Egypt
| | - Arwa Ali
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia.,Medical Oncology Department, South Egypt Cancer Institute, Assiut University, Assiut 71515, Egypt
| | - Mai Abdelgelil
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia.,Clinical Oncology Department, Assiut University Hospital, Assiut 71515, Egypt
| | - Esam Murshid
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia
| | - Abdulaziz Alhamad
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia
| | - Muhammad Ali
- Department of Oncology, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia.,Department of Radiation Oncology, Icon Cancer Centre, Warrnambool, Victoria 3280, Australia
| | - Hidayath Ansari
- Department of Imaging, Cleveland Clinic Abu Dhabi, Abu Dhabi 112412, United Arab Emirates
| | - Syed Hussain
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - John Glaholm
- Department of Oncology, Royal Marsden Hospital, London SW3 6JJ, UK
| |
Collapse
|
20
|
Thakur A, Ke X, Chen YW, Motallebnejad P, Zhang K, Lian Q, Chen HJ. The mini player with diverse functions: extracellular vesicles in cell biology, disease, and therapeutics. Protein Cell 2021; 13:631-654. [PMID: 34374936 PMCID: PMC9233731 DOI: 10.1007/s13238-021-00863-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/08/2021] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicles (EVs) are tiny biological nanovesicles ranging from approximately 30-1000 nm in diameter that are released into the extracellular matrix of most cell types and in biofluids. The classification of EVs includes exosomes, microvesicles, and apoptotic bodies, dependent on various factors such as size, markers, and biogenesis pathways. The transition of EV relevance from that of being assumed as a trash bag to be a key player in critical physiological and pathological conditions has been revolutionary in many ways. EVs have been recently revealed to play a crucial role in stem cell biology and cancer progression via intercellular communication, contributing to organ development and the progression of cancer. This review focuses on the significant research progress made so far in the role of the crosstalk between EVs and stem cells and their niche, and cellular communication among different germ layers in developmental biology. In addition, it discusses the role of EVs in cancer progression and their application as therapeutic agents or drug delivery vehicles. All such discoveries have been facilitated by tremendous technological advancements in EV-associated research, especially the microfluidics systems. Their pros and cons in the context of characterization of EVs are also extensively discussed in this review. This review also deliberates the role of EVs in normal cell processes and disease conditions, and their application as a diagnostic and therapeutic tool. Finally, we propose future perspectives for EV-related research in stem cell and cancer biology.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA.,The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Xiaoshan Ke
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA.,The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Ya-Wen Chen
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, 90089, USA.,Department of Stem Cell Biology and Regenerative Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - Pedram Motallebnejad
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA.,The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Kui Zhang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA.,The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Qizhou Lian
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong. .,Prenatal Diagnostic Center and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. .,HKUMed Laboratory of Cellular Therapeutics, the University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Huanhuan Joyce Chen
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA. .,The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
21
|
Liu Y, Zhang J, Du J, Song K, Liu J, Wang X, Li B, Ouyang R, Miao Y, Sun Y, Li Y. Biodegradable BiOCl platform for oxidative stress injury-enhanced chemodynamic/radiation therapy of hypoxic tumors. Acta Biomater 2021; 129:280-292. [PMID: 34033970 DOI: 10.1016/j.actbio.2021.05.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022]
Abstract
Various physiological characteristics of the tumor microenvironment (TME), such as hypoxia, overexpression of glutathione (GSH) and hydrogen peroxide (H2O2), and mild acidity, can severely reduce the efficacy of many cancer therapies. Altering the redox balance of the TME and increasing oxidative stress can accordingly enhance the efficacy of tumor therapy. Herein, we developed a bismuth-based Cu2+-doped BiOCl nanotherapeutic platform, BCHN, able to self-supply H2O2 for TME-regulated chemodynamic therapy (CDT) combined with sensitized radiotherapy (RT). BCHN released H2O2 and consumed GSH to degrade the composite in the slightly acidic TME, and generated hydroxyl radicals (•OH) via a Fenton-like reaction catalyzed by copper ions, to achieve oxidative stress-enhanced CDT. The Fenton-like reaction also catalyzed H2O2 to produce O2 to relieve tumor hypoxia, and combined with the X-ray-blocking property of bismuth to realize TME-enhanced radiotherapy. Synergistic CDT/RT has previously been shown to effectively inhibit tumor cell proliferation and achieve effective tumor control. The current results demonstrated a highly efficient multifunctional bio-degradable nanoplatform for oncotherapy. STATEMENT OF SIGNIFICANCE: Tumor microenvironment-modulated synergy of radiotherapy and chemodynamic therapy is conducive to rapid tumor ablation. Based on this principle, we fabricated a biodegradable BiOCl-based nanocomposite, BCHN. By supplying H2O2, a Fenton-like reaction generated •OH and O2 catalyzed by copper ions, and consumed glutathione to biodegrade the composite. Overall, these actions increased tumor oxidative stress and realized the synergistic anti-tumor actions of chemodynamic therapy combined with bismuth-based sensitization radiotherapy. This strategy thus provides a unique approach to oncology therapy.
Collapse
|
22
|
Checker R, Patwardhan RS, Jayakumar S, Maurya DK, Bandekar M, Sharma D, Sandur SK. Chemical and biological basis for development of novel radioprotective drugs for cancer therapy. Free Radic Res 2021; 55:595-625. [PMID: 34181503 DOI: 10.1080/10715762.2021.1876854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ionizing radiation (IR) causes chemical changes in biological systems through direct interaction with the macromolecules or by causing radiolysis of water. This property of IR is harnessed in the clinic for radiotherapy in almost 50% of cancers patients. Despite the advent of stereotactic radiotherapy instruments and other advancements in shielding techniques, the inadvertent deposition of radiation dose in the surrounding normal tissue can cause late effects of radiation injury in normal tissues. Radioprotectors, which are chemical or biological agents, can reduce or mitigate these toxic side-effects of radiotherapy in cancer patients and also during radiation accidents. The desired characteristics of an ideal radioprotector include low chemical toxicity, high risk to benefit ratio and specific protection of normal cells against the harmful effects of radiation without compromising the cytotoxic effects of IR on cancer cells. Since reactive oxygen species (ROS) are the major contributors of IR mediated toxicity, plethora of studies have highlighted the potential role of antioxidants to protect against IR induced damage. However, owing to the lack of any clinically approved radioprotector against whole body radiation, researchers have shifted the focus toward finding alternate targets that could be exploited for the development of novel agents. The present review provides a comprehensive insight in to the different strategies, encompassing prime molecular targets, which have been employed to develop radiation protectors/countermeasures. It is anticipated that understanding such factors will lead to the development of novel strategies for increasing the outcome of radiotherapy by minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Sundarraj Jayakumar
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Mayuri Bandekar
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| |
Collapse
|
23
|
Gezer A, Karadag-Sari E. The role of amifostine in preventing radiotherapy induced testicular tissue damage in rats. Biotech Histochem 2021; 97:215-221. [PMID: 34058938 DOI: 10.1080/10520295.2021.1933178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The germinal epithelium of the adult testis is susceptible to radiation induced damage. Amifostine is a drug used to prevent the side effects of radiotherapy (RT) and chemotherapy. We investigated the protective role of amifostine against RT induced damage to rat testis using the TUNEL assay. We used adult male rats divided equally into four groups: untreated control group; amifostine group, 200 mg/kg amifostine/day for 3 days; RT-saline group, 2 Gy/day local irradiation of testes for 3 days; RT-amifostine group, 2 Gy/day local irradiation of testes for 3 days plus 200 mg/kg amifostine 30 min before each irradiation. Four weeks after treatment, rats were sacrificed for histological examination and apoptosis was assessed using the TUNEL method. The TUNEL staining density was obtained by evaluating separate seminiferous tubules selected randomly from each section using the stereological fractionator method. Apoptosis in the seminiferous tubules in the control group and amifostine groups were evaluated as spontaneous. Frequent apoptosis was observed in the RT-saline group; a statistically significant difference was observed between the RT treated and untreated groups. Administration of amifostine 30 min before RT protected the testicular germ cells against apoptosis.
Collapse
Affiliation(s)
- Arzu Gezer
- Vocational School of Health Services, Ataturk University, Erzurum, Turkey
| | - Ebru Karadag-Sari
- Histology Department, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| |
Collapse
|
24
|
Shin SW, Yang K, Lee M, Moon J, Son A, Kim Y, Choi S, Kim DH, Choi C, Lee N, Park HC. Manganese Ferrite Nanoparticles Enhance the Sensitivity of Hepa1-6 Hepatocellular Carcinoma to Radiation by Remodeling Tumor Microenvironments. Int J Mol Sci 2021; 22:ijms22052637. [PMID: 33807943 PMCID: PMC7961905 DOI: 10.3390/ijms22052637] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
We evaluated the effect of manganese ferrite nanoparticles (MFN) on radiosensitization and immunologic responses using the murine hepatoma cell line Hepa1-6 and the syngeneic mouse model. The clonogenic survival of Hepa1-6 cells was increased by hypoxia, while being restricted by ionizing radiation (IR) and/or MFN. Although MFN suppressed HIF-1α under hypoxia, the combination of IR and MFN enhanced apoptosis and DNA damage in Hepa1-6 cells. In the Hepa1-6 syngeneic mouse model, the combination of IR and MFN notably limited the tumor growth compared to the single treatment with IR or MFN, and also triggered more frequent apoptosis in tumor tissues than that observed under other conditions. Increased expression of PD-L1 after IR was not observed with MFN alone or the combination of IR and MFN in vitro and in vivo, and the percentage of tumor-infiltrating T cells and cytotoxic T cells increased with MFN, regardless of IR, in the Hepa1-6 syngeneic mouse model, while IR alone led to T cell depletion. MFN might have the potential to overcome radioresistance by alleviating hypoxia and strengthening antitumor immunity in the tumor microenvironment.
Collapse
Affiliation(s)
- Sung-Won Shin
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
- Department of Medicine, Samsung Medical Center, Sungkyunwan University School of Medicine, Seoul 06351, Korea
| | - Kyungmi Yang
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
- Department of Medicine, Samsung Medical Center, Sungkyunwan University School of Medicine, Seoul 06351, Korea
| | - Miso Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul 02707, Korea; (M.L.); (J.M.)
| | - Jiyoung Moon
- School of Advanced Materials Engineering, Kookmin University, Seoul 02707, Korea; (M.L.); (J.M.)
| | - Arang Son
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
| | - Yeeun Kim
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
| | - Suha Choi
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
| | - Do-hyung Kim
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
| | - Changhoon Choi
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul 02707, Korea; (M.L.); (J.M.)
- Correspondence: (N.L.); (H.C.P.)
| | - Hee Chul Park
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Korea; (S.-W.S.); (K.Y.); (A.S.); (Y.K.); (S.C.); (D.-h.K.); (C.C.)
- Department of Medicine, Samsung Medical Center, Sungkyunwan University School of Medicine, Seoul 06351, Korea
- Correspondence: (N.L.); (H.C.P.)
| |
Collapse
|
25
|
Onder GO, Balcioglu E, Baran M, Ceyhan A, Cengiz O, Suna PA, Yıldız OG, Yay A. The different doses of radiation therapy-induced damage to the ovarian environment in rats. Int J Radiat Biol 2021; 97:367-375. [PMID: 33320730 DOI: 10.1080/09553002.2021.1864497] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE The sequelae of premature loss of ovarian function can undoubtedly have undesirable effects for a woman although radiotherapy is one of the most relevant treatment modalities for various types of malignancies. The aim of this study was to determine the effect of different doses of radiation on ovarian folliculogenesis, inflammation, and apoptotic markers. MATERIALS AND METHODS For this purpose, 40 healthy Wistar albino female rats divided into four groups: 1) Control group; 2) those that were exposed to total body 1 Gy of gamma rays; 3) those that were exposed to the total body 5 Gy of gamma rays, and 4) those that were exposed to total body 10 Gy of gamma rays. External irradiation to the total body was given with gamma irradiation delivered by the Co60 teletherapy machine. The day after radiation application the rats were sacrificed and the ovaries were removed in all groups. Histopathologic examination, follicle counting, and classification were performed in the ovarian tissues. The expression of AMH, TNF-α, IL1-β, Bax, and Bcl-2 was detected. The stained sections were examined for caspase 3 positive apoptotic cell numbers. RESULTS The recorded results revealed that increased radiation dose induced obvious ovarian injuries that were indicated by histopathological, and immunohistochemical alterations, including elevation of ovarian injury markers. A significantly lower number of total and primordial follicles was detected with increasing radiation dose compared with the control group. According to our immunohistochemical results, 10 Gy of gamma rays group had the lowest AMH expression levels, while had the highest TNF-α, IL1-β expression level compared to the control group. When the groups were evaluated in terms of apoptosis, it was seen that the number of caspase 3 positive cells and Bax immunoreactivity intensity increased with radiation dose. In contrast, Bcl-2 immunoreactivity intensity decreased with increasing radiation dose compared with the control group. CONCLUSIONS We demonstrate here that dose rate plays an important role when estimating the relation between exposure to an increased dose of ionizing radiation and the risk of ovarian disease. According to these results, certain factors have to be optimized before introducing them into clinics.
Collapse
Affiliation(s)
- Gozde Ozge Onder
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Esra Balcioglu
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Munevver Baran
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Ayse Ceyhan
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Ozge Cengiz
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Pinar Alisan Suna
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Oguz Galip Yıldız
- Department of Radiation Oncology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Arzu Yay
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| |
Collapse
|
26
|
Kalendralis P, Eyssen D, Canters R, Luk SM, Kalet AM, van Elmpt W, Fijten R, Dekker A, Zegers CM, Bermejo I. External validation of a Bayesian network for error detection in radiotherapy plans. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2021. [DOI: 10.1109/trpms.2021.3070656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Petros Kalendralis
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands. (e-mail: )
| | - Denis Eyssen
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| | - Richard Canters
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| | - Samuel M.H. Luk
- Department of Radiation Oncology, University of Washington Medical Center, Seattle, WA 98195-6043, USA
| | - Alan M. Kalet
- Department of Radiation Oncology, University of Washington Medical Center, Seattle, WA 98195-6043, USA
| | - Wouter van Elmpt
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| | - Rianne Fijten
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| | - Catharina M.L. Zegers
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| | - Inigo Bermejo
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands
| |
Collapse
|
27
|
Sun W, Bergmeier AP, Liao Y, Wu S, Tong L. CIRP Sensitizes Cancer Cell Responses to Ionizing Radiation. Radiat Res 2021; 195:93-100. [PMID: 33429432 PMCID: PMC8969209 DOI: 10.1667/rade-20-00063.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 10/05/2020] [Indexed: 11/03/2022]
Abstract
Cold inducible RNA binding protein (CIRP), also named A18 hnRNP or CIRBP, is a cold-shock RNA-binding protein which can be induced upon various cellular stresses. Its expression level is induced in various cancer tissues relative to adjacent normal tissues; this is believed to play a critical role in cancer development and progression. In this study, we investigated the role of CIRP in cells exposed to ionizing radiation. Our data show that CIRP reduction causes cell colony formation and cell viability reduction after irradiation. In addition, CIRP knockdown cells demonstrated a higher DNA damage rate but less cell cycle arrest after irradiation. As a result, the induced DNA damage with less DNA repair processes led to an increased cell apoptosis rate in CIRP knockdown cells postirradiation. These findings suggest that CIRP is a critical protein in irradiated cells and can be used as a potential target for sensitizing cancer cells to radiation therapy.
Collapse
Affiliation(s)
- Weichao Sun
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Adele P. Bergmeier
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biological Sciences and Ohio University, Ohio 45701
| | - Yi Liao
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shiyong Wu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Chemistry and Biochemistry and Ohio University, Ohio 45701
- Program of Molecular and Cellular Biology, Ohio University, Ohio 45701
| | - Lingying Tong
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Chemistry and Biochemistry and Ohio University, Ohio 45701
| |
Collapse
|
28
|
Lenarczyk M, Laiakis EC, Mattson DL, Johnson BD, Kronenberg A, North PE, Komorowski R, Mäder M, Baker JE. Irradiation of the kidneys causes pathologic remodeling in the nontargeted heart: A role for the immune system. FASEB Bioadv 2020; 2:705-719. [PMID: 33336158 PMCID: PMC7734425 DOI: 10.1096/fba.2020-00071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiac disease is a frequent and significant adverse event associated with radiotherapy for cancer. Identifying the underlying mechanism responsible for radiation injury to the heart will allow interventions to be developed. In the present study, we tested if local kidney irradiation results in remodeling of the shielded, nontargeted heart. One kidney, two kidneys, or the total body of male WAG and Dahl SS rats were irradiated with 10 Gy of X-rays. Local kidney irradiation resulted in systemic hypertension, increased BUN, infiltration of T lymphocytes, natural killer cells, and macrophages into the renal cortex and medulla, and renal fibrosis. Local irradiation of kidneys in WAG rats resulted in remodeling in the nontargeted heart after 120 days, manifested by perivascular fibrosis and increased interventricular septal thickness, but was not seen in Dahl SS rats due to a high baseline level of fibrosis in the sham-irradiated animals. Genetic depletion of T cells mitigated the nephropathy after local kidney irradiation, indicating a role for the immune system in mediating this outcome. Local kidney irradiation resulted in a cascade of pro-inflammatory cytokines and low-molecular weight metabolites into the circulation associated with transmission of signals resulting in pathologic remodeling in the nontargeted heart. A new model is proposed whereby radiation-induced cardiac remodeling in susceptible animals is indirect, with lower hemi body organs such as the kidney exporting factors into the circulation that cause remodeling outside of the irradiated field in the shielded, nontargeted heart. This nontargeted effect appears to be mediated, in part, by the immune system.
Collapse
Affiliation(s)
| | | | | | | | - Amy Kronenberg
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | | | | | | | | |
Collapse
|
29
|
Shabeeb D, Musa AE, Abd Ali HS, Najafi M. Curcumin Protects Against Radiotherapy-Induced Oxidative Injury to the Skin. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3159-3163. [PMID: 32848362 PMCID: PMC7429408 DOI: 10.2147/dddt.s265228] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/24/2020] [Indexed: 01/16/2023]
Abstract
Objective Side-effects to normal tissues reduce the therapeutic window of radiotherapy. During radiotherapy, the skin is inevitably exposed to doses of ionizing radiation, leading to varying degrees of skin damage. Natural antioxidants have been explored for their radioprotective potentials. Thus, the present study aimed to investigate the protective effect of curcumin against radiotherapy-induced oxidative damage to the skin. Methods Forty rats were divided into four groups as follows: vehicle control (without irradiation or drug treatment), treatment with 150 mg/kg curcumin, 10 Gy single dose irradiation only, and 150 mg/kg curcumin plus 10 Gy radiation (RC). In the treatment groups, each rat was treated orally with 150 mg/kg curcumin 1 day before irradiation to 3 consecutive days after irradiation. Weeks 1, 2, or 4 after irradiation, all rats were sacrificed and their skin tissues collected and frozen at −80°C for the determination of malondialdehyde (MDA), catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activity in skin tissues. Results Radiotherapy-induced oxidative injury to the skin was evidenced by elevated MDA levels as well as depleted CAT, SOD, and GSH-Px activities. However, the administration of curcumin before and after irradiation prevented radiotherapy-induced oxidative damage by significantly elevating the activities of antioxidant enzymes. Conclusion From the findings of the present study, curcumin showed potential for protection against radiotherapy-induced oxidative injury to the skin. However, future studies are required to evaluate its clinical efficacy.
Collapse
Affiliation(s)
- Dheyauldeen Shabeeb
- Department of Physiology, School of Medicine, University of Misan, Misan, Iraq.,Misan Radiotherapy Center, Ministry of Health, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Masoud Najafi
- Department of Radiology and Nuclear Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
30
|
Patel P, Malipatlolla DK, Devarakonda S, Bull C, Rascón A, Nyman M, Stringer A, Tremaroli V, Steineck G, Sjöberg F. Dietary Oat Bran Reduces Systemic Inflammation in Mice Subjected to Pelvic Irradiation. Nutrients 2020; 12:nu12082172. [PMID: 32707913 PMCID: PMC7468988 DOI: 10.3390/nu12082172] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022] Open
Abstract
Patients undergoing radiotherapy to treat pelvic-organ cancer are commonly advised to follow a restricted fiber diet. However, reducing dietary fiber may promote gastrointestinal inflammation, eventually leading to deteriorated intestinal health. The goal of this study was to evaluate the influence of dietary fiber on radiation-induced inflammation. C57BL/6J male mice were fed a High-oat bran diet (15% fiber) or a No-fiber diet (0% fiber) and were either irradiated (32 Gy delivered in four fractions) to the colorectal region or only sedated (controls). The dietary intervention started at 2 weeks before irradiation and lasted for 1, 6, and 18 weeks after irradiation, at which time points mice were sacrificed and their serum samples were assayed for 23 cytokines and chemokines. Our analyses show that irradiation increased the serum cytokine levels at all the time points analyzed. The No-fiber irradiated mice had significantly higher levels of pro-inflammatory cytokines than the High-oat irradiated mice at all time points. The results indicate that a fiber-rich oat bran diet reduces the intensity of radiation-induced inflammation, both at an early and late stage. Based on the results, it seems that the advice to follow a low-fiber diet during radiotherapy may increase the risk of decreased intestinal health in cancer survivors.
Collapse
Affiliation(s)
- Piyush Patel
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden
- Correspondence:
| | - Dilip Kumar Malipatlolla
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Sravani Devarakonda
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Cecilia Bull
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Ana Rascón
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100 Lund, Sweden; (A.R.); (M.N.)
| | - Margareta Nyman
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100 Lund, Sweden; (A.R.); (M.N.)
| | - Andrea Stringer
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia;
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Gunnar Steineck
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Fei Sjöberg
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden
| |
Collapse
|
31
|
Datta NR, Kok HP, Crezee H, Gaipl US, Bodis S. Integrating Loco-Regional Hyperthermia Into the Current Oncology Practice: SWOT and TOWS Analyses. Front Oncol 2020; 10:819. [PMID: 32596144 PMCID: PMC7303270 DOI: 10.3389/fonc.2020.00819] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Moderate hyperthermia at temperatures between 40 and 44°C is a multifaceted therapeutic modality. It is a potent radiosensitizer, interacts favorably with a host of chemotherapeutic agents, and, in combination with radiotherapy, enforces immunomodulation akin to “in situ tumor vaccination.” By sensitizing hypoxic tumor cells and inhibiting repair of radiotherapy-induced DNA damage, the properties of hyperthermia delivered together with photons might provide a tumor-selective therapeutic advantage analogous to high linear energy transfer (LET) neutrons, but with less normal tissue toxicity. Furthermore, the high LET attributes of hyperthermia thermoradiobiologically are likely to enhance low LET protons; thus, proton thermoradiotherapy would mimic 12C ion therapy. Hyperthermia with radiotherapy and/or chemotherapy substantially improves therapeutic outcomes without enhancing normal tissue morbidities, yielding level I evidence reported in several randomized clinical trials, systematic reviews, and meta-analyses for various tumor sites. Technological advancements in hyperthermia delivery, advancements in hyperthermia treatment planning, online invasive and non-invasive MR-guided thermometry, and adherence to quality assurance guidelines have ensured safe and effective delivery of hyperthermia to the target region. Novel biological modeling permits integration of hyperthermia and radiotherapy treatment plans. Further, hyperthermia along with immune checkpoint inhibitors and DNA damage repair inhibitors could further augment the therapeutic efficacy resulting in synthetic lethality. Additionally, hyperthermia induced by magnetic nanoparticles coupled to selective payloads, namely, tumor-specific radiotheranostics (for both tumor imaging and radionuclide therapy), chemotherapeutic drugs, immunotherapeutic agents, and gene silencing, could provide a comprehensive tumor-specific theranostic modality akin to “magic (nano)bullets.” To get a realistic overview of the strength (S), weakness (W), opportunities (O), and threats (T) of hyperthermia, a SWOT analysis has been undertaken. Additionally, a TOWS analysis categorizes future strategies to facilitate further integration of hyperthermia with the current treatment modalities. These could gainfully accomplish a safe, versatile, and cost-effective enhancement of the existing therapeutic armamentarium to improve outcomes in clinical oncology.
Collapse
Affiliation(s)
- Niloy R Datta
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephan Bodis
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
32
|
Ganguly D, Chandra Santra R, Mazumdar S, Saha A, Karmakar P, Das S. Radioprotection of thymine and calf thymus DNA by an azo compound: mechanism of action followed by DPPH radical quenching & ROS depletion in WI 38 lung fibroblast cells. Heliyon 2020; 6:e04036. [PMID: 32490245 PMCID: PMC7262411 DOI: 10.1016/j.heliyon.2020.e04036] [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: 03/15/2020] [Revised: 04/26/2020] [Accepted: 05/18/2020] [Indexed: 11/19/2022] Open
Abstract
Purpose To explain the observed radio-protection properties of an azo compound, 2-(2-hydroxyphenylazo)-indole-3∕-acetic acid (HPIA). Materials and methods Mechanism of radioprotection by HPIA was attempted using the stable free radical 2, 2-diphenyl-1-picrylhydrazyl (DPPH) using UV-Vis and electron paramagnetic resonance (EPR) spectroscopy. The radical destroying ability of HPIA was studied by depletion of reactive oxygen species (ROS) in WI 38 lung fibroblast cells. Results & Discussion Studies indicate HPIA interacts with radical intermediates formed in solution following irradiation by 60Co γ-rays. As a result, reactive radical intermediates do not cause any damage on chosen substrates like thymine or calf thymus DNA when irradiated in presence of HPIA. The study showed that reactive intermediates not only react with HPIA but that the kinetics of their reaction is definitely faster than their interaction either with thymine or with DNA. Had this not been the case, much more damage would have been observed on chosen substrates following irradiation with 60Co γ-rays, in the presence of HPIA than actually observed in experiments, particularly those that were performed in a relatively high dose. Experiments reveal radiation induced-damage caused to thymine in presence of HPIA was ~ 136 to ~ 132times that caused in its absence under different conditions indicating the radio-protection properties of HPIA. In case of calf thymus DNA, damage in presence of HPIA was much lower than in its absence. A fluorometric microplate assay for depletion of ROS by detecting the oxidation of 2′,7′-dichlorofluorescin-diacetate (DCF-DA) into the highly fluorescent compound 2′,7′ dichlorofluorescein (DCF) indicated HPIA brought about a considerable check on ROS-mediated damage to cells by scavenging them right away. Conclusion The study indicates HPIA may be an antioxidant supplement during radiotherapy.
Collapse
Affiliation(s)
- Durba Ganguly
- Department of Chemistry (Inorganic Section), Jadavpur University, Kolkata 700032, India
| | - Ramesh Chandra Santra
- Department of Chemistry (Inorganic Section), Jadavpur University, Kolkata 700032, India
| | - Swagata Mazumdar
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Saha
- UGC-DAE CSR, Kolkata Centre, Sector III, LB- 8, Bidhan Nagar, Kolkata 700 098, India
| | - Parimal Karmakar
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Saurabh Das
- Department of Chemistry (Inorganic Section), Jadavpur University, Kolkata 700032, India
- Corresponding author.
| |
Collapse
|
33
|
Soko GF, Burambo AB, Mngoya MM, Abdul BA. Public Awareness and Perceptions of Radiotherapy and Their Influence on the Use of Radiotherapy in Dar es Salaam, Tanzania. J Glob Oncol 2020; 5:1-10. [PMID: 31702947 PMCID: PMC6882515 DOI: 10.1200/jgo.19.00175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Misconceptions associated with radiotherapy (RT) may affect the patient’s choice to undergo or not undergo RT. In this study, the level of awareness and perceptions about radiation and RT, as well as their impact on the use of RT, were assessed. MATERIALS AND METHODS A cross-sectional survey was conducted in the city of Dar es Salaam, Tanzania, between November 2018 and March 2019. Stratified sampling was used to obtain a sample of 629 participants from 4 strata, including 53 patients with cancer, 129 health professionals, 127 medical and nursing students, and 320 respondents from the general public. A questionnaire with 13 items measuring awareness and 8 items measuring perceptions was used for data collection. The Kruskal-Wallis test and χ2 test were used to test association between predictor and outcome variables. Statistical analyses were performed using statistical software. RESULTS The percentage of right responses was < 50% in all 13 awareness items. Only 16.9% of respondents were aware that RT would not reduce their lifespan. Only 34.5% of respondents had positive perceptions of RT. Awareness was higher among medical/nursing students, younger respondents, single or cohabiting respondents, and those who had attained a college or higher education. Overall, 52% would accept receiving RT if recommended as part of their treatment. Those who would accept undergoing RT were more likely to have higher awareness and a positive perception of RT. CONCLUSION Public awareness of RT in Dar es Salaam is low, and negative perceptions prevail. Low levels of awareness and negative perceptions have a negative effect on the use of RT.
Collapse
|
34
|
Vinorelbine Augments Radiotherapy in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12040872. [PMID: 32260169 PMCID: PMC7226273 DOI: 10.3390/cancers12040872] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022] Open
Abstract
There is a need to improve the effectiveness of radiotherapy (RT) in hepatocellular carcinoma (HCC). Therefore, the purpose of this study was to explore the efficacy and toxicity of the anti-microtubule agent Vinorelbine as a radiosensitizer in HCC. The radio sensitivity of 16 HCC patient-derived xenograft (PDX) models was determined by quantifying the survival fraction following irradiation in vitro, and Vinorelbine radio sensitization was determined by clonogenic assay. Ectopic HCC xenografts were treated with a single dose of 8 Gy irradiation and twice-weekly 3 mg/kg Vinorelbine. Tumor growth and changes in the proteins involved in DNA repair, angiogenesis, tumor cell proliferation, and survival were assessed, and the 3/16 (18.75%), 7/16 (43.75%), and 6/16 (37.5%) HCC lines were classified as sensitive, moderately sensitive, and resistant, respectively. The combination of RT and Vinorelbine significantly inhibited tumor growth, DNA repair proteins, angiogenesis, and cell proliferation, and promoted more apoptosis compared with RT or Vinorelbine treatment alone. Vinorelbine improved HCC tumor response to standard irradiation with no increase in toxicity. HCC is prevalent in less developed parts of the world and is mostly unresectable on presentation. Vinorelbine and conventional radiotherapy are cost-effective, well-established modalities of cancer treatment that are readily available. Therefore, this strategy can potentially address an unmet clinical need, warranting further investigation in early-phase clinical trials.
Collapse
|
35
|
Sia J, Szmyd R, Hau E, Gee HE. Molecular Mechanisms of Radiation-Induced Cancer Cell Death: A Primer. Front Cell Dev Biol 2020; 8:41. [PMID: 32117972 PMCID: PMC7031160 DOI: 10.3389/fcell.2020.00041] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Radiation therapy (RT) is responsible for at least 40% of cancer cures, however treatment resistance remains a clinical problem. There have been recent advances in understanding the molecular mechanisms of radiation-induced cell death. The type of cell death after radiation depends on a number of factors including cell type, radiation dose and quality, oxygen tension, TP53 status, DNA repair capacity, cell cycle phase at time of radiation exposure, and the microenvironment. Mitotic catastrophe (a pathway preceding cell death that happens in mitosis or as a consequence of aberrant mitotic progression) is the primary context of radiation-induced cell death in solid cancers, although in a small subset of cancers such as haematopoietic malignancies, radiation results in immediate interphase apoptosis, occurring within hours after exposure. There is intense therapeutic interest in using stereotactic ablative body radiotherapy (SABR), a precise, high-dose form of RT given in a small number of fractions, to prime the immune system for cancer cell killing, but the optimal radiation dose and fractionation remain unclear. Additionally, promising novel radiosensitisers targeting the cell cycle and DNA repair pathways are being trialled. In the context of the increasing use of SABR and such novel agents in the clinic, we provide an updated primer on the major types of radiation-induced cell death, focussing on their molecular mechanisms, factors affecting their initiation, and their implications on immunogenicity.
Collapse
Affiliation(s)
- Joseph Sia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Radoslaw Szmyd
- Children's Medical Research Institute, Sydney, NSW, Australia.,Sydney West Radiation Oncology Network, Sydney, NSW, Australia
| | - Eric Hau
- Sydney West Radiation Oncology Network, Sydney, NSW, Australia.,The University of Sydney, Sydney, NSW, Australia
| | - Harriet E Gee
- Sydney West Radiation Oncology Network, Sydney, NSW, Australia.,The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
36
|
Zhou X, Liu H, Zheng Y, Han Y, Wang T, Zhang H, Sun Q, Li Z. Overcoming Radioresistance in Tumor Therapy by Alleviating Hypoxia and Using the HIF-1 Inhibitor. ACS APPLIED MATERIALS & INTERFACES 2020; 12:4231-4240. [PMID: 31912727 DOI: 10.1021/acsami.9b18633] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Radiotherapy has been extensively used to treat cancer patients because it can effectively damage most solid tumors without penetration limits. A hypoxic microenvironment in solid tumors leads to severe radioresistance and expression of hypoxic inducible factor-1 (HIF-1), which results in poor efficacy of radiotherapy alone. Herein, we report the excellent efficacy of radiotherapy achieved using a new type of yolk-shell Cu2-xSe@PtSe (CSP) nanosensitizer functionalized with the HIF-1α inhibitor acriflavine (ACF). We prepare the CSP nanosensitizer through the interfacial redox reactions between chloroplatinic acid and Cu2-xSe nanoparticles (CS) and then functionalize the nanosensitizer with ACF through their electrostatic interactions. We show that the synthesized CSP nanosensitizer can arrest the cell cycle (i.e., at the gap 2/mitosis (G2/M) phases) of tumor cells to enhance their sensitivity to X-rays and decompose endogenous H2O2 into O2 to reduce hypoxia and increase the production of reactive oxygen species, which leads to severe damage to DNA double strands and apoptosis of tumor cells. We also show that the ACF on the surface of CSP nanoparticles can effectively reduce the expression of HIF-1α. All these effects lead to a low vascular endothelial growth factor, low density of microvessels in tumor, decreased cell proliferation, and increased cell apoptosis, which synergistically and drastically enhance the efficacy of radiotherapy. This work provides insights and guidance for developing novel nanosensitizers to enhance the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Xingguo Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Yanhui Zheng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Tingting Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Qiao Sun
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou 215123 , P. R. China
| |
Collapse
|
37
|
Metformin Protects the Rat Small Intestine Against Radiation Enteritis. Jundishapur J Nat Pharm Prod 2019. [DOI: 10.5812/jjnpp.67352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
38
|
Musa AE, Omyan G, Esmaely F, Shabeeb D. Radioprotective Effect of Hesperidin: A Systematic Review. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E370. [PMID: 31336963 PMCID: PMC6681345 DOI: 10.3390/medicina55070370] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 01/23/2023]
Abstract
Background and objectives: Ionizing radiation (IR) has been of immense benefit to man, especially for medical purposes (diagnostic imaging and radiotherapy). However, the risks of toxicity in healthy normal cells, leading to cellular damage as well as early and late side effects, have been major drawbacks. The aim of this study was to evaluate the radioprotective effect of hesperidin against IR-induced damage. Materials and Methods: The preferred reporting items for systematic reviews and meta-analyses (PRISMA) were applied in reporting this study. A search was conducted using the electronic databases PubMed, Scopus, Embase, Google Scholar, and www.ClinicalTrials.gov for information about completed or ongoing clinical trials. Results: From our search results, 24 studies involving rats, mice, and cultured human and animal cells were included. An experimental case-control design was used in all studies. The studies showed that the administration of hesperidin reduced oxidative stress and inflammation in all investigated tissues. Furthermore, it increased 30-day and 60-day survival rates and protected against DNA damage. The best radioprotection was obtained when hesperidin was administered before irradiation. Conclusions: The results of the included studies support the antioxidant, anti-inflammatory, and antiapoptotic abilities of hesperidin as a potential radioprotective agent against IR-induced damage. We recommend future clinical trials for more insights.
Collapse
Affiliation(s)
- Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (TUMS), Tehran 1416753955, Iran.
| | - Gilnaz Omyan
- Department of Physics, University of Guilan, Guilan 43714, Iran
- Radiotherapy and Oncology Research Center, Cancer Institute, TUMS, Tehran 1416753955, Iran
| | - Farid Esmaely
- Department of Medical Physics, Tehran University of Medical Sciences (TUMS), Tehran 1416753955, Iran
| | - Dheyauldeen Shabeeb
- Misan Radiotherapy Center, Misan Health Directorate, Ministry of Health/Environment, Misan 62010, Iraq
- Department of Physiology, College of Medicine, University of Misan, Misan 62010, Iraq
| |
Collapse
|
39
|
|
40
|
Lenarczyk M, Kronenberg A, Mäder M, North PE, Komorowski R, Cheng Q, Little MP, Chiang IH, LaTessa C, Jardine J, Baker JE. Age at Exposure to Radiation Determines Severity of Renal and Cardiac Disease in Rats. Radiat Res 2019; 192:63-74. [PMID: 31095446 PMCID: PMC10654917 DOI: 10.1667/rr15043.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Radiotherapy with sparsely ionizing photons is a cornerstone of successful cancer treatment. Age at time of exposure to radiation is known to influence biological outcomes for many end points. The effect of dose and age at exposure upon the occurrence of radiogenic cardiovascular disease is poorly understood. The goal of this work was to determine the response of maleWAG/RijCmcr rats at 6 months of age to gamma rays, and at 6 months or 6 weeks of age to X rays, using clinically relevant biomarkers of cardiovascular disease and kidney injury. Overall, there were significant radiation-induced effects on the levels of bicarbonate (P=0.0016), creatinine (P=0.0002), calcium (P = 0.0009), triglycerides (P = 0.0269) and blood urea nitrogen, albumin, protein, AST, alkaline phosphatase, total cholesterol and HDL (all P < 0.0001). Of those variables with a significant radiation-dose effect, there were significant modifications by age at time of exposure for bicarbonate (P = 0.0033), creatinine (P = 0.0015), AST (P = 0.0040), total cholesterol (P = 0.0006) and blood urea nitrogen, calcium, albumin, protein, alkaline phosphatase and HDL (all P < 0.0001). Cardiac perivascular collagen content was significantly increased in rats that were 8.0 Gy X-ray irradiated at 6 weeks of age (P < 0.047) but not at 6 months of age. While systemic blood pressure was elevated in both cohorts after 8.0 Gy X-ray irradiation (compared to agematched sham-irradiated controls), the magnitude of the increase above baseline was greater in the younger rats (P < 0.05). These findings indicate that dose and age at time of irradiation determine the timeline and severity of cardiac and renal injury.
Collapse
Affiliation(s)
- Marek Lenarczyk
- Division of Congenital Cardiac Surgery, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Amy Kronenberg
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States of America
| | - Marylou Mäder
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Paula E. North
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Richard Komorowski
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Qunli Cheng
- Division of Congenital Cardiac Surgery, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Mark P. Little
- Radiation Epidemiology Branch, National Cancer Institute, Bethesda, MD, United States of America
| | - I-Hung Chiang
- Collider-Accelerator Department, Brookhaven National Laboratory, Upton, NY, United States of America
| | - Chiara LaTessa
- Collider-Accelerator Department, Brookhaven National Laboratory, Upton, NY, United States of America
| | - James Jardine
- Biology Department, Brookhaven National Laboratory, Upton, NY, United States of America
| | - John E. Baker
- Division of Congenital Cardiac Surgery, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States of America
| |
Collapse
|
41
|
Musa AE, Shabeeb D. Radiation-Induced Heart Diseases: Protective Effects of Natural Products. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E126. [PMID: 31075882 PMCID: PMC6572037 DOI: 10.3390/medicina55050126] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/27/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVDs) account for the majority of deaths worldwide. Radiation-induced heart diseases (RIHD) is one of the side effects following exposure to ionizing radiation (IR). Exposure could be from various forms such as diagnostic imaging, radiotherapy for cancer treatment, as well as nuclear disasters and nuclear accidents. RIHD is mostly observed after radiotherapy for thoracic malignancies, especially left breast cancer. RIHD may affect the supply of blood to heart muscles, leading to an increase in the risk of heart attacks to irradiated persons. Due to its dose-limiting consequence, RIHD has a negative effect on the therapeutic efficacy of radiotherapy. Several methods have been proposed for protection against RIHD. In this paper, we review the use of natural products, which have shown promising results for protection against RIHD.
Collapse
Affiliation(s)
- Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (TUMS), International Campus, Tehran 1416753955, Iran.
- Research Center for Molecular and Cellular Imaging, TUMS, Tehran 1416753955, Iran.
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan 62010, Iraq.
| |
Collapse
|
42
|
Farhood B, Ghorbani M. Dose Calculation Accuracy of Radiotherapy Treatment Planning Systems in Out-of-Field Regions. J Biomed Phys Eng 2019; 9:133-136. [PMID: 31214518 PMCID: PMC6538908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 03/28/2018] [Indexed: 12/02/2022]
Affiliation(s)
- B. Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - M. Ghorbani
- Biomedical Engineering and Medical Physics Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Beaton L, Bandula S, Gaze MN, Sharma RA. How rapid advances in imaging are defining the future of precision radiation oncology. Br J Cancer 2019; 120:779-790. [PMID: 30911090 PMCID: PMC6474267 DOI: 10.1038/s41416-019-0412-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/23/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022] Open
Abstract
Imaging has an essential role in the planning and delivery of radiotherapy. Recent advances in imaging have led to the development of advanced radiotherapy techniques—including image-guided radiotherapy, intensity-modulated radiotherapy, stereotactic body radiotherapy and proton beam therapy. The optimal use of imaging might enable higher doses of radiation to be delivered to the tumour, while sparing normal surrounding tissues. In this article, we review how the integration of existing and novel forms of computed tomography, magnetic resonance imaging and positron emission tomography have transformed tumour delineation in the radiotherapy planning process, and how these advances have the potential to allow a more individualised approach to the cancer therapy. Recent data suggest that imaging biomarkers that assess underlying tumour heterogeneity can identify areas within a tumour that are at higher risk of radio-resistance, and therefore potentially allow for biologically focussed dose escalation. The rapidly evolving concept of adaptive radiotherapy, including artificial intelligence, requires imaging during treatment to be used to modify radiotherapy on a daily basis. These advances have the potential to improve clinical outcomes and reduce radiation-related long-term toxicities. We outline how recent technological advances in both imaging and radiotherapy delivery can be combined to shape the future of precision radiation oncology.
Collapse
Affiliation(s)
- Laura Beaton
- Cancer Institute, University College London, London, UK
| | - Steve Bandula
- Cancer Institute, University College London, London, UK.,NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK
| | - Mark N Gaze
- NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK
| | - Ricky A Sharma
- Cancer Institute, University College London, London, UK. .,NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
44
|
Ahmad SS, Crittenden MR, Tran PT, Kluetz PG, Blumenthal GM, Bulbeck H, Baird RD, Williams KJ, Illidge T, Hahn SM, Lawrence TS, Spears PA, Walker AJ, Sharma RA. Clinical Development of Novel Drug-Radiotherapy Combinations. Clin Cancer Res 2019; 25:1455-1461. [PMID: 30498095 PMCID: PMC6397668 DOI: 10.1158/1078-0432.ccr-18-2466] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/16/2018] [Accepted: 11/26/2018] [Indexed: 12/25/2022]
Abstract
Radiotherapy is a fundamental component of treatment for the majority of patients with cancer. In recent decades, technological advances have enabled patients to receive more targeted doses of radiation to the tumor, with sparing of adjacent normal tissues. There had been hope that the era of precision medicine would enhance the combination of radiotherapy with targeted anticancer drugs; however, this ambition remains to be realized. In view of this lack of progress, the FDA-AACR-ASTRO Clinical Development of Drug-Radiotherapy Combinations Workshop was held in February 2018 to bring together stakeholders and opinion leaders from academia, clinical radiation oncology, industry, patient advocacy groups, and the FDA to discuss challenges to introducing new drug-radiotherapy combinations to the clinic. This Perspectives in Regulatory Science and Policy article summarizes the themes and action points that were discussed. Intelligent trial design is required to increase the number of studies that efficiently meet their primary outcomes; endpoints to be considered include local control, organ preservation, and patient-reported outcomes. Novel approaches including immune-oncology or DNA-repair inhibitor agents combined with radiotherapy should be prioritized. In this article, we focus on how the regulatory challenges associated with defining a new drug-radiotherapy combination can be overcome to improve clinical outcomes for patients with cancer.
Collapse
Affiliation(s)
- Saif S Ahmad
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Marka R Crittenden
- Translational Radiation Research, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon; The Oregon Clinic, Portland, Oregon
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins University, Baltimore, Maryland
| | - Paul G Kluetz
- FDA Oncology Center of Excellence, Silver Spring, Maryland
| | | | - Helen Bulbeck
- CTRad, National Cancer Research Institute, London, United Kingdom
| | - Richard D Baird
- Cambridge Breast Cancer Research Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kaye J Williams
- Division of Pharmacy and Optometry, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Tim Illidge
- Division of Cancer Sciences, The University of Manchester, Manchester Academic Health Science Centre, NIHR Biomedical Research Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Stephen M Hahn
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Patricia A Spears
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | - Amanda J Walker
- Department of Radiation Oncology, Wentworth Douglass Hospital and Seacoast Cancer Center, Dover, New Hampshire.
| | - Ricky A Sharma
- Radiation Oncology, NIHR University College London Hospitals Biomedical Research Centre, University College London Cancer Institute, University College London, London, United Kingdom.
| |
Collapse
|
45
|
Dong Y, Song Z, Luo Y, Ma X. A new proposal of utilizing intraoperative electron radiation therapy on the surface of liver to prevent postoperative liver metastasis of pancreatic cancer. Med Hypotheses 2019; 126:15-19. [PMID: 31010492 DOI: 10.1016/j.mehy.2019.02.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/27/2019] [Indexed: 02/05/2023]
Abstract
Pancreatic cancer is a lethal cancer with high rate of liver metastasis worldwide, whereas its treatment choices are limited to a large extent. The limitation of current therapeutic strategies calls for an effective approach which can lower the postoperative liver metastasis rate in order to improve the overall prognosis and survival rate. Comprehensively considering the basic knowledge and clinical practice of tumor treatment worldwide, we proposed three points of hypotheses. Basically, the existing evidences indicated that tumor cells shedding from pancreatic cancer localized in the marginal liver preferentially through the Portal vein. Then, the percentage depth dose distribution of electron radiation is consistent with the marginal distribution of liver metastasis from pancreatic cancer. Based on the characteristics of liver metastasis of pancreatic cancer and the percentage depth dose of electron radiation, we provide a new propose of preventing postoperative liver metastasis in a way of prophylactic intraoperative electron radiation therapy on the surface of liver. Intraoperative electron radiation is relatively easy to control radiation dose and treatment area under direct vision, effectively inhibiting the metastasis and growth of cancer cells and preventing further deterioration of pancreatic cancer patients' condition. Therefore, this hypothesis has an important clinical significance for postoperative rehabilitation and improvement of patients' survival.
Collapse
Affiliation(s)
- Yiting Dong
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Zikuan Song
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yuling Luo
- West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
46
|
The increased adhesion of tumor cells to endothelial cells after irradiation can be reduced by FAK-inhibition. Radiat Oncol 2019; 14:25. [PMID: 30717801 PMCID: PMC6360706 DOI: 10.1186/s13014-019-1230-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/23/2019] [Indexed: 12/30/2022] Open
Abstract
Background Radiotherapy is administered in more than 60% of all solid tumors. Most patients are cured but a significant number develops local recurrences or distant metastases. The question arises if irradiation might influence the metastatic process. In the present study we examined whether the adhesion of glioblastoma or breast cancer cells to endothelial cells, an important step in metastasis, is affected by photon irradiation. Methods U-87 MG, U-373 MG and MDA-MB-231 cancer cells as well as primary human endothelial cells were irradiated with 0, 2, 4, or 8 Gy photons at a dose rate of 5 Gy/min. The adhesion of cancer cells to endothelial cells was tested either with the Vybrant based assay via fluorescent labelling or with an ibidi pump system able to mimic the physiological blood flow in vitro. In addition, the impact of FAK (focal adhesion kinase) inhibitor PF-573, 228 on the adhesion of non-irradiated and irradiated tumor cells was analyzed. Adhesion related and regulated proteins were analyzed by Western blotting. Results The cellular adhesion was increased after irradiation regardless of which cell type was irradiated. The FAK-inhibitor was able to reduce the adhesion of non-irradiated cells but also the irradiation-induced increase in adhesion of tumor cells to endothelium. Adhesion related proteins were enhanced after irradiation with 4 Gy or 8 Gy in both cells types. The increased adhesion after irradiation is accompanied by the phosphorylation of src (Y416), FAK (Y397) and increased expression of paxillin. Conclusion Irradiation with photons in therapeutic doses is able to enhance the interaction between tumor cells and endothelial cells and by that might influence important steps of the metastatic process.
Collapse
|
47
|
Shabeeb D, Najafi M, Musa AE, Keshavarz M, Shirazi A, Hassanzadeh G, Hadian MR, Samandari H. Biochemical and Histopathological Evaluation of the Radioprotective Effects of Melatonin Against Gamma Ray-Induced Skin Damage. Curr Radiopharm 2019; 12:72-81. [PMID: 30465519 DOI: 10.2174/1874471012666181120163250] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Radiotherapy is one of the treatment methods for cancers using ionizing radiations. About 70% of cancer patients undergo radiotherapy. Radiation effect on the skin is one of the main complications of radiotherapy and dose limiting factor. To ameliorate this complication, we used melatonin as a radioprotective agent due to its antioxidant and anti-inflammatory effects, free radical scavenging, improving overall survival after irradiation as well as minimizing the degree of DNA damage and frequency of chromosomal abrasions. METHODS Sixty male Wistar rats were randomly assigned to 4 groups: control (C), melatonin (M), radiation (R) and melatonin + radiation (MR). A single dose of 30 Gy gamma radiation was exposed to the right hind legs of the rats while 40 mg/ml of melatonin was administered 30 minutes before irradiation and 2 mg/ml once daily in the afternoon for one month till the date of rat's sacrifice. Five rats from each group were sacrificed 4, 12 and 20 weeks after irradiation. Afterwards, their exposed skin tissues were examined histologically and biochemically. RESULTS In biochemical analysis, we found that malondialdehyde (MDA) levels significantly increased in R group and decreased significantly in M and MR groups after 4, 12, and 20 weeks, whereas catalase (CAT) and superoxide dismutase (SOD) activities decreased in the R group and increased in M and MR groups during the same time periods compared with the C group (p<0.05). Histopathological examination found there were statistically significant differences between R group compared with the C and M groups for the three different time periods (p<0.005, p<0.004 and p<0.004) respectively, while R group differed significantly with MR group (p<0.013). No significant differences were observed between C and M compared with MR group (p>0.05) at 4 and 20 weeks except for inflammation and hair follicle atrophy, while there were significant effects at 12 weeks (p<0.05). CONCLUSION Melatonin can be successfully used for the prevention and treatment of radiation-induced skin injury. We recommend the use of melatonin in optimal and safe doses. These doses should be administered over a long period of time for effective radioprotection and amelioration of skin damages as well as improving the therapeutic ratio of radiotherapy.
Collapse
Affiliation(s)
- Dheyauldeen Shabeeb
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Science, International Campus, Tehran, Iran
- Department of Physiology, College of Medicine, University of Misan, Iraq
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Science, International Campus, Tehran, Iran
- Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansoor Keshavarz
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Science, International Campus, Tehran, Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammed Reza Hadian
- Brain and Spinal Cord Injury, Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Hedayat Samandari
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Le Fèvre C, Antoni D, Thiéry A, Noël G. Radiothérapie des métastases osseuses : revue multi-approches de la littérature. Cancer Radiother 2018; 22:810-825. [DOI: 10.1016/j.canrad.2017.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/20/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022]
|
49
|
Coffey M, Leech M. The European Society of Radiotherapy and Oncology (ESTRO) European Higher Education Area levels 7 and 8 postgraduate benchmarking document for Radiation TherapisTs (RTTs). Tech Innov Patient Support Radiat Oncol 2018; 8:22-40. [PMID: 32095587 PMCID: PMC7033774 DOI: 10.1016/j.tipsro.2018.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
This guideline details the European Higher Education Area Levels 7 and 8 Postgraduate benchmarking document for Radiation TherapisTs (RTTs). The purpose of this benchmarking document is to assist higher education institutes in the development of radiation therapy-specific curricula for RTTs engaging in postgraduate education, with a view to working at an advanced level in radiation therapy departments. The document specifies the knowledge, skills and competences that are required to work in specific areas of RTT practice, at levels 7 and 8. These include: advanced delineation and volume determination, advanced treatment planning, advanced imaging, quality and risk management, management and service development, patient care and support, brachytherapy and research.
Collapse
Affiliation(s)
- Mary Coffey
- Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College, Dublin 2, Ireland
| | - Michelle Leech
- Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College, Dublin 2, Ireland
| | | |
Collapse
|
50
|
Jin H, Ko YS, Kim HJ. P2Y2R-mediated inflammasome activation is involved in tumor progression in breast cancer cells and in radiotherapy-resistant breast cancer. Int J Oncol 2018; 53:1953-1966. [PMID: 30226596 PMCID: PMC6192788 DOI: 10.3892/ijo.2018.4552] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
In the tumor microenvironment, extracellular nucleotides are released and accumulate, and can activate the P2Y2 receptor (P2Y2R), which regulates various responses in tumor cells, resulting in tumor progression and metastasis. Moreover, the inflammasome has recently been reported to be associated with tumor progression. However, the role of P2Y2R in inflammasome activation in breast cancer cells is not yet well defined. Therefore, in this study, we investigated the role of P2Y2R in inflammasome-mediated tumor progression in breast cancer using breast cancer cells and radiotherapy-resistant (RT‑R) breast cancer cells. We established RT‑R-breast cancer cells (RT‑R‑MDA‑MB‑231, RT‑R‑MCF‑7, and RT‑R-T47D cells) by repeated irradiation (2 Gy each, 25 times) in a previous study. In this study, we found that the RT‑R breast cancer cells exhibited an increased release of adenosine triphosphate (ATP) and P2Y2R activity. In particular, the RT‑R‑MDA‑MB‑231 cells derived from highly metastatic MDA‑MB‑231 cells, exhibited a markedly increased ATP release, which was potentiated by tumor necrosis factor (TNF)-α. The MDA‑MB‑231 cells exhibited inflammasome activation, as measured by caspase‑1 activity and interleukin (IL)-1β secretion following treatment with TNF‑α and ATP; these effects were enhanced in the RT‑R‑MDA‑MB‑231 cells. However, the increased caspase‑1 activities and IL‑1β secretion levels induced in response to treatment with TNF‑α or ATP were significantly reduced by P2Y2R knockdown or the presence of apyrase in both the MDA‑MB‑231 and RT‑R‑MDA‑MB‑231 cells, suggesting the involvement of ATP-activated P2Y2R in inflammasome activation. In addition, TNF‑α and ATP increased the invasive and colony-forming ability of the MDA‑MB‑231 and RT‑R‑MDA‑MB‑231 cells, and these effects were caspase‑1-dependent. Moreover, matrix metalloproteinase (MMP)-9 activity was modulated by caspase-1, in a P2Y2R-dependent manner in the MDA‑MB‑231 and RT‑R‑MDA‑MB‑231 cells. Finally, nude mice injected with the RT‑R‑MDA‑MB‑231-EV cells (transfected with the empty vector) exhibited increased tumor growth, and higher levels of MMP-9 in their tumors and IL‑1β levels in their serum compared with the mice injected with the RT‑R‑MDA‑MB‑231-P2Y2R shRNA cells (transfected with P2Y2R shRNA). On the whole, the findings of this study suggest that extracellular ATP promotes tumor progression in RT‑R-breast cancer cells and breast cancer cells by modulating invasion and associated molecules through the P2Y2R-inflammasome activation pathway.
Collapse
Affiliation(s)
- Hana Jin
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, Gyeongsang 52727, Republic of Korea
| | - Young Shin Ko
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, Gyeongsang 52727, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, Gyeongsang 52727, Republic of Korea
| |
Collapse
|