1
|
Sutthiwanjampa C, Kang SH, Kim MK, Hwa Choi J, Kim HK, Woo SH, Bae TH, Kim WJ, Kang SH, Park H. Tumor necrosis factor-α-treated human adipose-derived stem cells enhance inherent radiation tolerance and alleviate in vivo radiation-induced capsular contracture. J Adv Res 2025; 72:433-449. [PMID: 39019109 DOI: 10.1016/j.jare.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024] Open
Abstract
INTRODUCTION Post-mastectomy radiotherapy plays a crucial role in breast cancer treatment but can lead to an inflammatory response causing soft tissue damage, particularly radiation-induced capsular contracture (RICC), impacting breast reconstruction outcomes. Adipose-derived stem cells (ADSCs), known for their regenerative potential via paracrine capacity, exhibit inherent radiotolerance. The influence of tumor necrosis factor-alpha (TNF-α) on ADSCs has been reported to enhance the paracrine effect of ADSCs, promoting wound healing by modulating inflammatory responses. OBJECTIVE This study investigates the potential of TNF-α-treated human ADSCs (T-hASCs) on silicone implants to alleviate RICC, hypothesizing to enhance suppressive effects on RICC by modulating inflammatory responses in a radiation-exposed environment. METHODS In vitro, T-hASCs were cultured on various surfaces to assess viability after exposure to radiation up to 20 Gy. In vivo, T-hASC and non-TNF-α-treated hASC (C-hASCs)-coated membranes were implanted in mice before radiation exposure, and an evaluation of the RICC mitigation took place 4 and 8 weeks after implantation. In addition, the growth factors released from T-hASCs were assessed. RESULTS In vitro, hASCs displayed significant radiotolerance, maintaining consistent viability after exposure to 10 Gy. TNF-α treatment further enhanced radiation tolerance, as evidenced by significantly higher viability than C-hASCs at 20 Gy. In vivo, T-hASC-coated implants effectively suppressed RICC, reducing capsule thickness. T-hASCs exhibited remarkable modulation of the inflammatory response, suppressing M1 macrophage polarization while enhancing M2 polarization. The elevated secretion of vascular endothelial growth factor from T-hASCs is believed to induce macrophage polarization, potentially reducing RICC. CONCLUSION This study establishes T-hASCs as a promising strategy for ameliorating the adverse effects experienced by breast reconstruction patients after mastectomy and radiation therapy. The observed radiotolerance, anti-fibrotic effects, and immune modulation suggest the possibility of enhancing patient outcomes and quality of life. Further research and clinical trials are warranted for broader clinical uses.
Collapse
Affiliation(s)
- Chanutchamon Sutthiwanjampa
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Seung Hyun Kang
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Mi Kyung Kim
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Departments of Pathology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Jin Hwa Choi
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Radiation Oncology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Han Koo Kim
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Soo Hyun Woo
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Tae Hui Bae
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Woo Joo Kim
- Department of Plastic Surgery, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong-si, Gyeonggi-do 14353, Republic of Korea
| | - Shin Hyuk Kang
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
2
|
Tayal S, Kaur N, Kaur T, Chadha VD. Zinc as an adjunct in radiation-based therapies: Evidences of radioprotection and mechanistic insights. Nutr Health 2025:2601060251329404. [PMID: 40388708 DOI: 10.1177/02601060251329404] [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: 05/21/2025]
Abstract
BackgroundRadiation-based therapies are a progressive modality for managing life-threatening diseases such as cancer. However, these treatments often inflict damage on non-target tissues, necessitating the development of effective radioprotective agents. Zinc, known for its diversified role under various pathological conditions, has emerged as a potential protective agent against radiation-induced injuries due to its antioxidant, anti-inflammatory and immune-regulating properties.AimThis review aims to evaluate the potential of zinc in mitigating the adverse effects associated with radiation-based therapies, focusing on its protective role in normal tissue injury.MethodsA comprehensive literature review was conducted using multiple databases, including PubMed, NCBI, SciFinder, Google Scholar and Science Direct. Relevant studies assessing the radioprotective effects of zinc were identified and analysed to summarise its efficacy and potential benefits in radiation therapy.ResultsThe review highlights the beneficial effects of zinc in managing radiation-induced adverse effects, such as oral mucositis, skin injury, dermatitis, xerostomia, dysgeusia, dysosmia, bone marrow regeneration and oxidative stress reduction. Zinc's role as an antioxidant and anti-inflammatory agent, along with its ability to regulate immune system homeostasis, underpins these protective effects.ConclusionsZinc shows promising potential as a radioprotective agent in mitigating the adverse effects of radiation-based therapies. Despite the positive preclinical and clinical findings, further randomised trials with larger sample sizes and rigorous methodologies are needed to confirm zinc's efficacy. Additionally, further research is warranted to explore zinc's potential in addressing other radiation-induced events, ultimately contributing to improved patient care during radiation therapy.
Collapse
Affiliation(s)
- Sachin Tayal
- Centre for Nuclear Medicine, Panjab University, Chandigarh, India
| | - Navpreet Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Vijayta D Chadha
- Centre for Nuclear Medicine, Panjab University, Chandigarh, India
| |
Collapse
|
3
|
Park SW, Shin J, Jeong BK, Byun S, Lee KS, Choi J. The Effects of Extracorporeal Shock Wave Therapy on Cutaneous Radiation Injury in a Mouse Model. Plast Reconstr Surg 2025; 155:813-825. [PMID: 39344004 DOI: 10.1097/prs.0000000000011782] [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] [Indexed: 10/01/2024]
Abstract
BACKGROUND Although radiation-induced skin injuries are a concern in patients receiving radiation therapy, there are few effective treatments. The aim of this study was to evaluate the protective effects of extracorporeal shock wave therapy (ESWT) on irradiated fibroblasts and mouse skin. METHODS In this in vitro study of human dermal fibroblasts, the experimental group was subjected to ESWT after irradiation (20 Gy). The control groups were only irradiated or only subjected to ESWT. At 24 or 48 hours after ESWT, cell viability, cell migration, and mRNA and protein expression were measured. In the in vivo study, the experimental group (7 mice) was treated with ESWT after irradiation (45 Gy). The control group (7 mice) was only irradiated. At 8 weeks after irradiation, dorsal skin was harvested for histopathologic examination and protein isolation. RESULTS In dermal fibroblasts, treatment with ESWT increased viability of irradiated cells compared with irradiated-only and untreated cells ( P = 0.005). ESWT increased cell migration 24 hours after irradiation ( P = 0.002) and decreased transforming growth factor-β (TGF-β) protein expression 48 hours after irradiation ( P = 0.024). In mice, ESWT decreased the level of radiation-related skin injury ( P = 0.006). Treatment of irradiated skin with ESWT decreased TGF-β1 ( P = 0.009) and phospho-Smad3 ( P = 0.009) protein expression, decreased myofibroblasts ( P = 0.047), and increased vessel density ( P < 0.001). CONCLUSION This study demonstrated that ESWT alleviated radiation-induced fibrosis by downregulating TGF-β1 expression, suggesting the potential of ESWT for the treatment of radiation-induced fibrosis.
Collapse
Affiliation(s)
- Sang Woo Park
- From the Department of Plastic and Reconstructive Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine
| | - Jaebong Shin
- Department of Plastic and Reconstructive Surgery, Gyeongsang National University School of Medicine
| | - Bae Kwon Jeong
- Department of Radiation Oncology, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital
| | | | - Kyung Suk Lee
- Department of Plastic and Reconstructive Surgery, Gyeongsang National University School of Medicine
| | - Jaehoon Choi
- Plastic and Reconstructive Surgery, Keimyung University School of Medicine
| |
Collapse
|
4
|
Xu M, Peng Q, Zhang J, Xu Z, Cheng X, Cao Z, Zhang Y. Comparative Transcriptomic Analysis Unveils Divergent Effects of FLASH Versus Conventional Irradiation on Skin Cells. Dose Response 2025; 23:15593258251342837. [PMID: 40401244 PMCID: PMC12092997 DOI: 10.1177/15593258251342837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/23/2025] Open
Abstract
Objectives FLASH radiotherapy is garnering attention for its capacity to diminish skin toxicity without compromising tumoricidal efficacy, presenting a stark contrast to conventional (CONV) radiotherapy. Despite its promise, the underlying molecular mechanisms of FLASH irradiation (FLASH-IR) on skin are not yet fully elucidated. Methods This study investigated the transcriptomic responses of human foreskin fibroblast cells (HFF-1) via the FLASH-IR or CONV irradiation (CONV-IR), employing the next-generation RNA sequencing (RNA-seq) to capture the gene expression profiles. Our comparative analysis aimed to dissect the cellular and molecular pathways influenced by these two irradiation methods. Results We identified a spectrum of differentially expressed genes (DEGs), signaling pathways, and transcriptional networks that were either shared or divergent between FLASH-IR and CONV-IR. Particularly, transcription factor NR4A1 showed significant upregulation in response to FLASH-IR, while chromatin stability factor ELF3 was markedly downregulated following CONV-IR. The top 10 up-regulated DEGs were subjected to qPCR validation, confirming their differential expression in response to FLASH-IR and CONV-IR. Conclusion Collectively, our findings delineate unique regulatory landscapes of FLASH-IR and CONV-IR on skin cells, corroborating established effects and shedding new light on the molecular interplay within the context of ultra-high dose radiation.
Collapse
Affiliation(s)
- Mengmeng Xu
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Zhang
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiming Xu
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinyang Cheng
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhifei Cao
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongsheng Zhang
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Karasik D, Michaelson G, Cabrera CI, Nahra A, Zhao NW. Disparities in Dysphagia Care Among Head and Neck Cancer Patients: A Retrospective Cohort Study. OTO Open 2025; 9:e70090. [PMID: 40224296 PMCID: PMC11986681 DOI: 10.1002/oto2.70090] [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: 11/25/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 04/15/2025] Open
Abstract
Objective To investigate the incidence of dysphagia among head and neck cancer (HNC) patients and assess disparities in utilization of speech-language pathology (SLP) services across different demographic groups. Study Design Retrospective cohort study. Setting Analysis of data from the TriNetX global health network, comprising over 125 million deidentified electronic health records worldwide. Methods HNC patients diagnosed with oral, oropharyngeal, laryngeal, or nasopharyngeal cancer with and without dysphagia between January 1, 2004 and October 30, 2024 were identified. Patients were divided into two cohorts for comparison: those who received SLP services after dysphagia diagnosis and those who did not. The association of demographic characteristics (sex, ethnicity, and race) with SLP services were analyzed. Results Of 269,629 HNC patients, 28.8% (n = 77,562) were diagnosed with dysphagia. Significant disparities were found: female and non-White patients were less likely to be diagnosed with dysphagia. Once diagnosed, female, Hispanic/Latino, and non-White patients were also significantly less likely to receive SLP services compared to female, Hispanic/Latino, and non-White patients. Overall, only 38.8% of patients with dysphagia received SLP services. Conclusion This study highlights significant sex, ethnic, and racial disparities in dysphagia diagnosis and SLP service utilization among HNC patients. Furthermore, SLP services are underutilized. There is a need for targeted interventions to increase dysphagia prevention and surveillance and ensure equitable access to dysphagia care, improving outcomes for all HNC survivors.
Collapse
Affiliation(s)
- Daniel Karasik
- Department of Otolaryngology–Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Case Western Reserve University School of MedicineClevelandOhioUSA
| | - Gillian Michaelson
- Department of Otolaryngology–Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Case Western Reserve University School of MedicineClevelandOhioUSA
| | - Claudia I. Cabrera
- Department of Otolaryngology–Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Case Western Reserve University School of MedicineClevelandOhioUSA
| | - Alexis Nahra
- Department of Otolaryngology–Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
| | - Nina W. Zhao
- Department of Otolaryngology–Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Case Western Reserve University School of MedicineClevelandOhioUSA
| |
Collapse
|
6
|
Vinasco-Sandoval T, Moratille S, Crechet F, Mesloub Y, Montanari J, Auvré F, Deleuze JF, Foray N, Fortunel NO, Martin MT. Long Noncoding VIM-AS1: Biomarker of Breast Fibrosis Susceptibility After Radiation Therapy and Promoter of Transforming Growth Factor Beta1-Driven Fibrosis. Int J Radiat Oncol Biol Phys 2025; 121:783-797. [PMID: 39436333 DOI: 10.1016/j.ijrobp.2024.09.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024]
Abstract
PURPOSE Fibrosis is a common late complication of radiation therapy. Molecular dysregulations leading to fibrosis have been characterized for the coding part of the genome, notably those involving the TGFB1 gene network. However, because a large part of the human genome encodes RNA transcripts that are not translated into proteins, exploring the involvement of the noncoding part of the genome in fibrosis susceptibility and development was the aim of this work. METHODS AND MATERIALS Breast cancer patients having or not having developed severe breast fibrosis after radiation therapy were retrospectively selected from the COPERNIC collection. Exome sequencing and RNA-seq transcriptomic profiling were performed on 19 primary dermal fibroblast strains isolated from the patients' nonirradiated skin. Functional experiments were based on fibrogenic induction by transforming growth factor-Beta1 (TGFB1) and gene knockdown in healthy donor fibroblasts. RESULTS Coding and noncoding transcriptomes discriminated fibrosis from nonfibrosis conditions, and a signature of breast fibrosis susceptibility comprising 15 long noncoding RNAs (lncRNAs) was identified. A hazard ratio validation showed that the lncRNA vimentin antisense long noncoding RNA 1 (VIM-AS1) was the best biomarker associated with fibrosis risk. This lncRNA has not been previously associated with any fibrotic disorder, but we found it upregulated in data sets from cardiac fibrosis and scleroderma, suggesting a general role in tissue fibrosis. Functional experiments demonstrated a profibrotic action of VIM-AS1 because its knockdown reduced myofibroblast activation, collagen matrix production, and dermal organoid contraction. RNA-seq data analysis after VIM-AS1 silencing also pointed out the regulation of replication, cell cycle, and DNA repair. Mechanistically, because VIM-AS1 was found coregulated with the vimentin gene, these data support a profibrotic function of the TGFB1/VIM-AS1/vimentin axis, targeting the dynamics of fibroblast-myofibroblast transition. CONCLUSIONS Noncoding RNA analysis can provide specific biomarkers relevant to the prediction of normal tissue responses after radiation therapy, which opens perspectives of next-generation approaches for treatment, in the frame of the recent developments of RNA-based technologies.
Collapse
Affiliation(s)
- Tatiana Vinasco-Sandoval
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France
| | - Sandra Moratille
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France
| | - Françoise Crechet
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France
| | - Yasmina Mesloub
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France
| | - Juliette Montanari
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France
| | - Frederic Auvré
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France
| | - Jean-François Deleuze
- CEA, Institut de Biologie François Jacob (IBFJ), Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Nicolas Foray
- INSERM, U1296 Unit, Radiation: Defense, Health and Environment, Centre Léon-Bérard, Lyon, France
| | - Nicolas O Fortunel
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France.
| | - Michele T Martin
- CEA, Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse (LGRK), Evry, France; Université Paris-Saclay, France; CEA, Institut de Biologie François Jacob (IBFJ), Département de Radiobiologie Cellulaire et Moléculaire (DRCM), Fontenay-aux-Roses, France.
| |
Collapse
|
7
|
Verginadis II, Citrin DE, Ky B, Feigenberg SJ, Georgakilas AG, Hill-Kayser CE, Koumenis C, Maity A, Bradley JD, Lin A. Radiotherapy toxicities: mechanisms, management, and future directions. Lancet 2025; 405:338-352. [PMID: 39827884 DOI: 10.1016/s0140-6736(24)02319-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 01/22/2025]
Abstract
For over a century, radiotherapy has revolutionised cancer treatment. Technological advancements aim to deliver high doses to tumours with increased precision while minimising off-target effects to organs at risk. Despite advancements such as image-guided, high-precision radiotherapy delivery, long-term toxic effects on healthy tissues remain a great clinical challenge. In this Review, we summarise common mechanisms driving acute and long-term side-effects and discuss monitoring strategies for radiotherapy survivors. We explore ways to mitigate toxic effects through novel technologies and proper patient selection and counselling. Additionally, we address policies and management strategies to minimise the severity and impact of toxicity during and after treatment. Finally, we examine the potential advantages of emerging technologies and innovative approaches to improve conformity, accuracy, and minimise off-target effects.
Collapse
Affiliation(s)
- Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bonnie Ky
- Department of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J Feigenberg
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Christine E Hill-Kayser
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Cancer Center, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Maity
- Department of Radiation Oncology, University of Utah Health, Salt Lake City, UT, USA
| | - Jeffrey D Bradley
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander Lin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Khateeb S. Etoricoxib-NLC Mitigates Radiation-Induced Ovarian Damage in Rats: Insights into Pro-Inflammatory Cytokines, Antioxidant Activity, and Hormonal Responses. Biomolecules 2024; 15:12. [PMID: 39858407 PMCID: PMC11761947 DOI: 10.3390/biom15010012] [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: 11/13/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Radiotherapy is a critical treatment for cancer but poses significant risks to ovarian tissue, particularly in young females, leading to premature ovarian failure (POF). This study examines the therapeutic potential of etoricoxib nanostructured lipid carriers (ETO-NLC) in mitigating radiation-induced ovarian damage in female Wistar rats. Twenty-four female rats were randomly assigned to four groups: a control group receiving normal saline, a group exposed to a single dose of whole-body gamma radiation (6 Gy), a group treated with etoricoxib (10 mg/kg) post-radiation, and a group treated with ETO-NLC for 14 days following radiation. Histopathological evaluations and oxidative stress biomarker assessments were conducted, including ELISAs for reactive oxygen species (ROS), pro-inflammatory cytokines (IL-1β, TNF-α), and signaling molecules (PI3K, AKT, P38MAPK, AMH). Serum levels of estrogen, FSH, and LH were measured, and gene expression analysis for TGF-β and Nrf2 was performed using qRT-PCR. The findings indicate that ETO-NLC has the potential to ameliorate the harmful effects of ovarian damage induced by γ-radiation. These therapeutic effects were achieved through the modulation of oxidative stress, inflammation, augmentation of antioxidant defenses (including Nrf2 activation), support for cell survival pathways (via PI3K/Akt signaling), regulation of MAPK, mitigation of fibrosis (TGF-β), and preservation of ovarian reserve (as evidenced by AMH, FSH/LH, and estrogen levels). ETO-NLC shows promise as an effective strategy for attenuating radiation-induced ovarian damage, highlighting the need for further research to enhance therapeutic interventions aimed at preserving ovarian function during cancer treatment.
Collapse
Affiliation(s)
- Sahar Khateeb
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Biochemistry Division, Department of Chemistry, Faculty of Science, Fayoum University, Fayoum P.O. Box 63514, Egypt;
| |
Collapse
|
9
|
Pouladvand N, Azarnia M, Zeinali H, Fathi R, Tavana S. An overview of different methods to establish a murine premature ovarian failure model. Animal Model Exp Med 2024; 7:835-852. [PMID: 39219374 PMCID: PMC11680483 DOI: 10.1002/ame2.12477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/14/2024] [Indexed: 09/04/2024] Open
Abstract
Premature ovarian failure (POF)is defined as the loss of normal ovarian function before the age of 40 and is characterized by increased gonadotropin levels and decreased estradiol levels and ovarian reserve, often leading to infertility. The incomplete understanding of the pathogenesis of POF is a major impediment to the development of effective treatments for this disease, so the use of animal models is a promising option for investigating and identifying the molecular mechanisms involved in POF patients and developing therapeutic agents. As mice and rats are the most commonly used models in animal research, this review article considers studies that used murine POF models. In this review based on the most recent studies, first, we introduce 10 different methods for inducing murine POF models, then we demonstrate the advantages and disadvantages of each one, and finally, we suggest the most practical method for inducing a POF model in these animals. This may help researchers find the method of creating a POF model that is most appropriate for their type of study and suits the purpose of their research.
Collapse
Affiliation(s)
- Negar Pouladvand
- Department of Embryology, Reproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
- Department of Animal Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Mahnaz Azarnia
- Department of Animal Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Hadis Zeinali
- Department of Animal Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
| | - Somayeh Tavana
- Department of Embryology, Reproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
| |
Collapse
|
10
|
Li K, Ji M, Sun X, Shan J, Su G. Food Polyphenols in Radiation-Related Diseases: The Roles and Possible Mechanisms. Curr Nutr Rep 2024; 13:884-895. [PMID: 39340730 DOI: 10.1007/s13668-024-00582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
PURPOSE OF REVIEW As science and technology continue to evolve, the potential harm of radiation to the human body cannot be overlooked. Radiation has the capacity to inflict cellular and body-wide damage. Polyphenols are a group of naturally occurring compounds that are found in an array of plant foods. Scientific studies have demonstrated that these compounds possess noteworthy anti-radiation efficacy. Furthermore, they have been observed to be less toxic at higher doses. In the present review, we discussed the mechanisms of ionizing radiation damage and the progress in the research on the radiation resistance mechanism of polyphenol compounds, to provide guidance for the prevention and treatment of radiation related diseases. RECENT FINDINGS Food polyphenols can reduce the oxidative damage caused by ionizing radiation, clear free radicals, reduce DNA damage, regulate NF-KB, MAPK, JAK/STAT, Wnt and other signaling pathways, improve immune function, and have significant protective effects on radiation-induced inflammation, fibrosis, cancer and other aspects. In addition, it also has significant dual effects on radiation sensitization and radiation protection. Food polyphenols come from a wide range of sources, are abundant in daily food, and have no toxic side effects, demonstrating that food polyphenols have great advantages in preventing and treating radiation-related diseases.
Collapse
Affiliation(s)
- Kaidi Li
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Maxin Ji
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiujuan Sun
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Junyan Shan
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Guangyue Su
- Shenyang Pharmaceutical University, Shenyang, 110016, China.
- Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning Province, Key Laboratory for TCM Material Basis Study and Innovative, Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
11
|
Pang N, Yang Z, Zhang W, Du Y, Zhang L, Li X, Peng Y, Qi X. Cancer-associated fibroblasts barrier breaking via TGF-β blockade paved way for docetaxel micelles delivery to treat pancreatic cancer. Int J Pharm 2024; 665:124706. [PMID: 39277152 DOI: 10.1016/j.ijpharm.2024.124706] [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: 05/29/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
TGF-β is a crucial regulator in tumor microenvironment (TME), especially for myofibroblastic cancer-associated fibroblasts (myCAFs). The myCAFs can be motivated by TGF-β signaling to erect pro-tumor TME, meanwhile, myCAFs overexpress TGF-β to mediate the crosstalk between tumor and stromal cells. The blockade of TGF-β can break cancer-associated fibroblasts barrier, consequently opening the access for drugs into tumor. The TGF-β is a promising target in anti-tumor therapy. Herein, we introduced a two-stage combination therapy (TC-Therapy), including TGF-β receptor I inhibitor SB525334 (SB) and cytotoxicity agent docetaxel micelle (DTX-M). We found that SB and DTX-M synergistically inhibited myCAFs proliferation and elevated p53 protein expression in BxPC-3/3T3 mixed cells. Gene and protein tests demonstrated that SB cut off TGF-β signaling via receptor blockade and it did not arouse TGF-β legend compensated internal autocrine. On the contrary, two agents combined decreased TGF-β secretion and inhibited myCAFs viability marked by α-SMA and FAPα. TC-Therapy was applied in BxPc-3/3T3 mixed tumor-bearing mice model. After TC-Therapy, the α-SMA+/ FAPα+ myCAFs faded increasingly and collagenous fibers mainly secreted by myCAFs decreased dramatically as well. More than that, the myCAFs barrier breaking helped to normalize micro-vessels and paved way for micelle penetration. The TGF-β protein level of TC-Therapy in TME was much lower than that of simplex DTX-M, which might account for TME restoration. In conclusion, TGF-β inhibitor acted as the pioneer before nano chemotherapeutic agents. The TC-Therapy of TGF-β signaling inhibition and anti-tumor agent DTX-M is a promising regimen without arising metastasis risk to treat pancreatic cancer. The therapeutic regimen focused on TGF-β related myCAFs reminds clinicians to have a comprehensive understanding of pancreatic cancer.
Collapse
Affiliation(s)
- Ning Pang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Zhenzhen Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenjie Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yitian Du
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lu Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xin Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiwei Peng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xianrong Qi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
12
|
Nisar A, Bin Zafar ST, Mistry A, Mohammad H. Giant paediatric thymoma and its tailored anaesthetic management. BMJ Case Rep 2024; 17:e260052. [PMID: 39532334 DOI: 10.1136/bcr-2024-260052] [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: 11/16/2024] Open
Abstract
A toddler presented with significant weight loss and lethargy. His CT scan chest revealed a sizeable anterior mediastinal mass of 14.5×12.0×7.0 cm, resulting in compression of the airway. The patient underwent a successful tumour resection but postoperatively experienced left diaphragmatic paralysis, which was effectively managed through conservative measures. Histopathology was consistent with thymoma (type B2, Masaoka stage I). This case emphasises the critical role of collaborative efforts among oncology, surgery and anaesthesiology teams in achieving positive outcomes for rare paediatric mediastinal tumours. It underscores the importance of comprehensive preoperative anaesthetic assessments, careful planning for potential complications and vigilant postoperative monitoring. Healthcare professionals, particularly anaesthesiologists, can find valuable insights in this case report for navigating the complexities associated with managing giant mediastinal masses in the paediatric population.
Collapse
Affiliation(s)
- Abdullah Nisar
- Department of Anesthesia, The Aga Khan University Hospital, Karachi, Pakistan
| | | | - Akbar Mistry
- Department of Anesthesia, The Aga Khan University Hospital, Karachi, Pakistan
| | - Hamid Mohammad
- Department of Anesthesia, The Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
13
|
Jang SY, Ahn IY, Bae TH, Kang SH, Woo SH, Kim WJ, Kim MK, Sutthiwanjampa C, Kim HK. Comparison of Effects of Acellular Dermal Matrix and Latissimus Dorsi Muscle Flap on Radiation-induced Peri-implant Capsular Contracture in a Rabbit Model. Arch Plast Surg 2024; 51:581-591. [PMID: 39544515 PMCID: PMC11560322 DOI: 10.1055/a-2368-1813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 07/13/2024] [Indexed: 11/17/2024] Open
Abstract
Background Capsular contracture of breast implants is a major complication in breast surgery. Clinically, covering a breast implant with acellular dermal matrix (ADM) or autologous tissue is considered to be the most effective technique to prevent capsular contracture. This study was designed to compare the protective effects of ADM and latissimus dorsi (LD) muscle flap placement on capsular contracture by increasing the rate of capsular contracture through controlled radiation exposure in a rabbit model. Methods Twenty New Zealand white rabbits were divided into three groups. After the implant was placed beneath the pectoralis major muscle, the lateral third of the implant was left exposed in the control group ( n = 6). In the ADM group ( n = 7), the exposed implant was covered with AlloDerm. In the LD flap group ( n = 7), the exposed implant was covered with a pedicled LD muscle flap. All groups were irradiated 3 weeks after implant insertion. After 6 months, peri-implant tissues were harvested and analyzed. Results ADM showed markedly lower myofibroblast activity than the LD flap. However, transforming growth factor-β1 levels and the activity of collagen types I and III produced in fibroblasts were significantly lower in the ADM group than in the LD flap group. Conclusion Based on the findings of our rabbit experiments, ADM is expected to have a comparative advantage in reducing the risk of capsular contracture compared to the LD flap.
Collapse
Affiliation(s)
- Suk Yoon Jang
- Department of Plastic and Reconstructive Surgery, Chung-Ang University, Graduate School of Medicine, Seoul, Republic of Korea
| | - Il Young Ahn
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Tae Hui Bae
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Republic of Korea
| | - Shin Hyuk Kang
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Soo Hyun Woo
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Woo Ju Kim
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Republic of Korea
| | - Mi Kyung Kim
- Department of Pathology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | | | - Han Koo Kim
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
14
|
Nunez-Alvarez L, Ledwon JK, Applebaum S, Progri B, Han T, Laudo J, Tac V, Gosain AK, Tepole AB. Tissue expansion mitigates radiation-induced skin fibrosis in a porcine model. Acta Biomater 2024; 189:427-438. [PMID: 39326692 PMCID: PMC11570334 DOI: 10.1016/j.actbio.2024.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Tissue expansion (TE) is the primary method for breast reconstruction after mastectomy. In many cases, mastectomy patients undergo radiation treatment (XR). Radiation is known to induce skin fibrosis and is one of the main causes for complications during post-mastectomy breast reconstruction. TE, on the other hand, induces a pro-regenerative response that culminates in growth of new skin. However, the combined effect of XR and TE on skin mechanics is unknown. Here we used the porcine model of TE to study the effect of radiation on skin fibrosis through biaxial testing, histological analysis, and kinematic analysis of skin deformation over time. We found that XR leads to stiffening of skin compared to control based on a shift in the transition stretch (transition between a low stiffness and an exponential stress-strain region characteristic of collagenous tissue) and an increase in the high modulus (modulus computed with stress-stretch data past the transition point). The change in transition stretch can be explained by thicker, more aligned collagen fiber bundles measured in histology images. Skin subjected to both XR+TE showed similar microstructure to controls as well as similar biaxial response, suggesting that physiological remodeling of collagen induced by TE partially counteracts pro-fibrotic XR effects. Skin growth was indirectly assessed with a kinematic approach that quantified increase in permanent area changes without reduction in thickness, suggesting production of new tissue driven by TE even in the presence of radiation treatment. Future work will focus on the detailed biological mechanisms by which TE counteracts radiation induced fibrosis. STATEMENT OF SIGNIFICANCE: Breast cancer is the most prevalent in women and its treatment often results in total breast removal (mastectomy), followed by reconstruction using tissue expanders. Radiation, which is used in about a third of breast reconstruction cases, can lead to significant complications. The timing of radiation treatment remains controversial. Radiation is known to cause immediate skin damage and long-term fibrosis. Tissue expansion leads to a pro-regenerative response involving collagen remodeling. Here we show that tissue expansion immediately prior to radiation can reduce the level of radiation-induced fibrosis. Thus, we anticipate that this new evidence will open up new avenues of investigation into how the collagen remodeling and pro-regenerative effects of tissue expansion can be leverage to prevent radiation-induced fibrosis.
Collapse
Affiliation(s)
| | | | | | | | - Tianhong Han
- School of Mechanical Engineering, Purdue University United States
| | - Joel Laudo
- School of Mechanical Engineering, Purdue University United States
| | - Vahidullah Tac
- School of Mechanical Engineering, Purdue University United States
| | - Arun K Gosain
- Lurie Children's Hospital United States; Department of Plastic and Reconstructive Surgery, Northwestern School of Medicine United States
| | - Adrian Buganza Tepole
- Weldon School of Biomedical Engineering, Purdue University United States; School of Mechanical Engineering, Purdue University United States.
| |
Collapse
|
15
|
Cui J, Wang TJ, Zhang YX, She LZ, Zhao YC. Molecular biological mechanisms of radiotherapy-induced skin injury occurrence and treatment. Biomed Pharmacother 2024; 180:117470. [PMID: 39321513 DOI: 10.1016/j.biopha.2024.117470] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024] Open
Abstract
Radiotherapy-Induced Skin Injury (RISI) is radiation damage to normal skin tissue that primarily occurs during tumor Radiotherapy and occupational exposure. The risk of RISI is high due to the fact that the skin is not only the first body organ that ionizing radiation comes into contact with, but it is also highly sensitive to it, especially the basal cell layer and capillaries. Typical clinical manifestations of RISI include erythema, dry desquamation, moist desquamation, and ulcers, which have been established to significantly impact patient care and cancer treatment. Notably, our current understanding of RISI's pathological mechanisms and signaling pathways is inadequate, and no standard treatments have been established. Radiation-induced oxidative stress, inflammatory responses, fibrosis, apoptosis, and cellular senescence are among the known mechanisms that interact and promote disease progression. Additionally, radiation can damage all cellular components and induce genetic and epigenetic changes, which play a crucial role in the occurrence and progression of skin injury. A deeper understanding of these mechanisms and pathways is crucial for exploring the potential therapeutic targets for RISI. Therefore, in this review, we summarize the key mechanisms and potential treatment methods for RISI, offering a reference for future research and development of treatment strategies.
Collapse
Affiliation(s)
- Jie Cui
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Tie-Jun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Yu-Xuan Zhang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Li-Zhen She
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Yue-Chen Zhao
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| |
Collapse
|
16
|
Touny AA, Venkataraman B, Ojha S, Pessia M, Subramanian VS, Hariharagowdru SN, Subramanya SB. Phytochemical Compounds as Promising Therapeutics for Intestinal Fibrosis in Inflammatory Bowel Disease: A Critical Review. Nutrients 2024; 16:3633. [PMID: 39519465 PMCID: PMC11547603 DOI: 10.3390/nu16213633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVE Intestinal fibrosis, a prominent consequence of inflammatory bowel disease (IBD), presents considerable difficulty owing to the absence of licensed antifibrotic therapies. This review assesses the therapeutic potential of phytochemicals as alternate methods for controlling intestinal fibrosis. Phytochemicals, bioactive molecules originating from plants, exhibit potential antifibrotic, anti-inflammatory, and antioxidant activities, targeting pathways associated with inflammation and fibrosis. Compounds such as Asperuloside, Berberine, and olive phenols have demonstrated potential in preclinical models by regulating critical signaling pathways, including TGF-β/Smad and NFκB, which are integral to advancing fibrosis. RESULTS The main findings suggest that these phytochemicals significantly reduce fibrotic markers, collagen deposition, and inflammation in various experimental models of IBD. These phytochemicals may function as supplementary medicines to standard treatments, perhaps enhancing patient outcomes while mitigating the adverse effects of prolonged immunosuppressive usage. Nonetheless, additional clinical trials are necessary to validate their safety, effectiveness, and bioavailability in human subjects. CONCLUSIONS Therefore, investigating phytochemicals may lead to crucial advances in the formulation of innovative treatment approaches for fibrosis associated with IBD, offering a promising avenue for future therapeutic development.
Collapse
Affiliation(s)
- Aya A. Touny
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Ahram Canadian University, Giza 12581, Egypt
| | - Balaji Venkataraman
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Mauro Pessia
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
| | | | - Shamanth Neralagundi Hariharagowdru
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sandeep B. Subramanya
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
17
|
Lawrence J, Seelig D, Demos-Davies K, Ferreira C, Ren Y, Wang L, Alam SK, Yang R, Guedes A, Craig A, Hoeppner LH. Radiation dermatitis in the hairless mouse model mimics human radiation dermatitis. Sci Rep 2024; 14:24819. [PMID: 39438583 PMCID: PMC11496547 DOI: 10.1038/s41598-024-76021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Over half of all people diagnosed with cancer receive radiation therapy. Moderate to severe radiation dermatitis occurs in most human radiation patients, causing pain, aesthetic distress, and a negative impact on tumor control. No effective prevention or treatment for radiation dermatitis exists. The lack of well-characterized, clinically relevant animal models of human radiation dermatitis contributes to the absence of strategies to mitigate radiation dermatitis. Here, we establish and characterize a hairless SKH-1 mouse model of human radiation dermatitis by correlating temporal stages of clinical and pathological skin injury. We demonstrate that a single ionizing radiation treatment of 30 Gy using 6 MeV electrons induces severe clinical grade 3 peak toxicity at 12 days, defined by marked erythema, desquamation and partial ulceration, with resolution occurring by 25 days. Histopathology reveals that radiation-induced skin injury features temporally unique inflammatory changes. Upregulation of epidermal and dermal TGF-ß1 and COX-2 protein expression occurs at peak dermatitis, with sustained epidermal TGF-ß1 expression beyond resolution. Specific histopathological variables that remain substantially high at peak toxicity and early clinical resolution, including epidermal thickening, hyperkeratosis and dermal fibroplasia/fibrosis, serve as specific measurable parameters for in vivo interventional preclinical studies that seek to mitigate radiation-induced skin injury.
Collapse
Affiliation(s)
- Jessica Lawrence
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, 1365 Gortner Ave, St Paul, MN, 55108, USA.
- Masonic Cancer Center, University of Minnesota, 425 East River Parkway, Minneapolis, MN, 55455, USA.
- Department of Radiation Oncology, Medical School, University of Minnesota, 516 Delaware St SE, Minneapolis, MN, 55455, USA.
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA.
| | - Davis Seelig
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, 1365 Gortner Ave, St Paul, MN, 55108, USA
- Masonic Cancer Center, University of Minnesota, 425 East River Parkway, Minneapolis, MN, 55455, USA
| | - Kimberly Demos-Davies
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, 1365 Gortner Ave, St Paul, MN, 55108, USA
| | - Clara Ferreira
- Department of Radiation Oncology, Medical School, University of Minnesota, 516 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Yanan Ren
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
- Department of Urology, Northwestern University, 303 E Superior Street, Chicago, IL, 60611, USA
| | - Li Wang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
| | - Rendong Yang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
- Department of Urology, Northwestern University, 303 E Superior Street, Chicago, IL, 60611, USA
| | - Alonso Guedes
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, 1365 Gortner Ave, St Paul, MN, 55108, USA
| | - Angela Craig
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, 1365 Gortner Ave, St Paul, MN, 55108, USA
- Masonic Cancer Center, University of Minnesota, 425 East River Parkway, Minneapolis, MN, 55455, USA
- Hennepin Healthcare Research Institute, 701 Park Ave, Suite S3, Minneapolis, MN, 55415, USA
| | - Luke H Hoeppner
- Masonic Cancer Center, University of Minnesota, 425 East River Parkway, Minneapolis, MN, 55455, USA.
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| |
Collapse
|
18
|
Muñoz Forti K, Weisman GA, Jasmer KJ. Cell type-specific transforming growth factor-β (TGF-β) signaling in the regulation of salivary gland fibrosis and regeneration. J Oral Biol Craniofac Res 2024; 14:257-272. [PMID: 38559587 PMCID: PMC10979288 DOI: 10.1016/j.jobcr.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/13/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024] Open
Abstract
Salivary gland damage and hypofunction result from various disorders, including autoimmune Sjögren's disease (SjD) and IgG4-related disease (IgG4-RD), as well as a side effect of radiotherapy for treating head and neck cancers. There are no therapeutic strategies to prevent the loss of salivary gland function in these disorders nor facilitate functional salivary gland regeneration. However, ongoing aquaporin-1 gene therapy trials to restore saliva flow show promise. To identify and develop novel therapeutic targets, we must better understand the cell-specific signaling processes involved in salivary gland regeneration. Transforming growth factor-β (TGF-β) signaling is essential to tissue fibrosis, a major endpoint in salivary gland degeneration, which develops in the salivary glands of patients with SjD, IgG4-RD, and radiation-induced damage. Though the deposition and remodeling of extracellular matrix proteins are essential to repair salivary gland damage, pathological fibrosis results in tissue hardening and chronic salivary gland dysfunction orchestrated by multiple cell types, including fibroblasts, myofibroblasts, endothelial cells, stromal cells, and lymphocytes, macrophages, and other immune cell populations. This review is focused on the role of TGF-β signaling in the development of salivary gland fibrosis and the potential for targeting TGF-β as a novel therapeutic approach to regenerate functional salivary glands. The studies presented highlight the divergent roles of TGF-β signaling in salivary gland development and dysfunction and illuminate specific cell populations in damaged or diseased salivary glands that mediate the effects of TGF-β. Overall, these studies strongly support the premise that blocking TGF-β signaling holds promise for the regeneration of functional salivary glands.
Collapse
Affiliation(s)
- Kevin Muñoz Forti
- Christopher S. Bond Life Sciences Center and Department of Biochemistry, University of Missouri, United States
| | - Gary A. Weisman
- Christopher S. Bond Life Sciences Center and Department of Biochemistry, University of Missouri, United States
| | - Kimberly J. Jasmer
- Christopher S. Bond Life Sciences Center and Department of Biochemistry, University of Missouri, United States
| |
Collapse
|
19
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
20
|
Fijardo M, Kwan JYY, Bissey PA, Citrin DE, Yip KW, Liu FF. The clinical manifestations and molecular pathogenesis of radiation fibrosis. EBioMedicine 2024; 103:105089. [PMID: 38579363 PMCID: PMC11002813 DOI: 10.1016/j.ebiom.2024.105089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 04/07/2024] Open
Abstract
Advances in radiation techniques have enabled the precise delivery of higher doses of radiotherapy to tumours, while sparing surrounding healthy tissues. Consequently, the incidence of radiation toxicities has declined, and will likely continue to improve as radiotherapy further evolves. Nonetheless, ionizing radiation elicits tissue-specific toxicities that gradually develop into radiation-induced fibrosis, a common long-term side-effect of radiotherapy. Radiation fibrosis is characterized by an aberrant wound repair process, which promotes the deposition of extensive scar tissue, clinically manifesting as a loss of elasticity, tissue thickening, and organ-specific functional consequences. In addition to improving the existing technologies and guidelines directing the administration of radiotherapy, understanding the pathogenesis underlying radiation fibrosis is essential for the success of cancer treatments. This review integrates the principles for radiotherapy dosimetry to minimize off-target effects, the tissue-specific clinical manifestations, the key cellular and molecular drivers of radiation fibrosis, and emerging therapeutic opportunities for both prevention and treatment.
Collapse
Affiliation(s)
- Mackenzie Fijardo
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Yin Yee Kwan
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States of America
| | - Kenneth W Yip
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Fei-Fei Liu
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
21
|
Pilśniak A, Szlauer-Stefańska A, Tukiendorf A, Rutkowski T, Składowski K, Kamińska-Winciorek G. Dermoscopy of Chronic Radiation-Induced Dermatitis in Patients with Head and Neck Cancers Treated with Radiotherapy. Life (Basel) 2024; 14:399. [PMID: 38541724 PMCID: PMC10971316 DOI: 10.3390/life14030399] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2025] Open
Abstract
Radiotherapy (RT) is an integral part of many cancer treatment protocols. Chronic radiation-induced dermatitis (CRD) is a cutaneous toxicity that occurs in one-third of all patients treated with this method. CRD is usually observed several months after completion of treatment. Typical symptoms of CRD are telangiectasia, skin discoloration, atrophy, thickening, and cutaneous fibrosis. There are currently no data in the literature on the evaluation of the dermoscopic features of CRD. The aim of this prospective study was the identification of clinical and dermoscopic features in a group of 32 patients with head and neck cancer (HNC) in whom CRD developed after RT. CRD was assessed at 3, 6, and 12 months after RT in 16, 10, and 10 patients, respectively. CRD was assessed at one time point and two time points in 28 and 4 patients, respectively. The control included skin areas of the same patient not exposed to RT. The dataset consisted of 36 clinical and 216 dermoscopic photos. Clinical evaluation was performed according to the RTOG/EORTC radiation-induced dermatitis scale. The highest score was grade 2 observed in 21 patients. Clinical observations revealed the presence of slight and patchy atrophy, pigmentation change, moderate telangiectasias, and some and total hair loss. Dotted vessels, clustered vessel distribution, white patchy scale, perifollicular white color, white structureless areas, brown dots and globules, and white lines were the most frequently noted features in dermoscopy. Three independent risk factors for chronic toxicity, such as age, gender, and surgery before RT, were identified. The dermoscopic features that had been shown in our study reflect the biological reaction of the skin towards radiation and may be used for the parametrization of CRD regarding its intensity and any other clinical consequences in the future.
Collapse
Affiliation(s)
- Aleksandra Pilśniak
- Department of Internal Medicine, Autoimmune and Metabolic Diseases, Faculty of Medical Sciences, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Anastazja Szlauer-Stefańska
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), 44-102 Gliwice, Poland;
| | | | - Tomasz Rutkowski
- Inpatient Department of Radiation and Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Gliwice Branch, 44-102 Gliwice, Poland; (T.R.); (K.S.)
| | - Krzysztof Składowski
- Inpatient Department of Radiation and Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), Gliwice Branch, 44-102 Gliwice, Poland; (T.R.); (K.S.)
| | - Grażyna Kamińska-Winciorek
- Department of Bone Marrow Transplantation and Onco-Hematology, Skin Cancer and Melanoma Team, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), 44-102 Gliwice, Poland
| |
Collapse
|
22
|
Cheng SW, Leung KHV, Mok KCJ, Yeung KW, Wong SYI, Lam YL, Ip KM, Lok YW, Wong ACL. Improvement in Swallowing Function in Patients with Previous Irradiation for Nasopharyngeal Carcinoma by Expiratory Muscle Strength Training. Dysphagia 2024; 39:129-139. [PMID: 37392211 DOI: 10.1007/s00455-023-10600-2] [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: 09/22/2022] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Dysphagia and chronic aspiration are common post-irradiation complications in nasopharyngeal carcinoma (NPC) survivors. Expiratory Muscle Strength Training (EMST) is a simple device-driven exercise therapy for swallowing training. This study investigates the effectiveness of EMST in a group of post-irradiated NPC patients. This prospective cohort, including twelve patients with previous irradiation for NPC and with swallowing disturbance, was performed between 2019 and 2021 in a single institution. Patients were trained with EMST for 8 weeks. Non-parametric analyses examined effects of EMST on primary outcome, maximum expiratory pressure. Secondary outcomes were measured with Penetration-aspiration scale, Yale pharyngeal residue severity rating scale (YPRSRS) by flexible endoscopic evaluation of swallowing, and Eating Assessment Tool (EAT-10) and M.D. Anderson Dysphagia Inventory questionnaire. Twelve patients, with a mean (SD) age of 64.3 (8.2) were recruited. There was no patient dropout with 88.9% overall compliance of training. Maximum expiratory pressure improved by 41% (median 94.5 to 133.5 cmH2O, p = 0.003). There was reduction in Penetration-aspiration scale with thin liquid (median 4 to 3, p = 0.026), and in YPRSRS at pyriform fossa with mildly thick liquid (p = 0.021) and at vallecula with thin liquid (p = 0.034), mildly thick liquid (p = 0.014) and pureed meat congee (p = 0.016). Questionnaire scores did not significantly change statistically. EMST is an easy-to-use and effective exercise therapy to improve airway safety and swallowing function in post-irradiated NPC survivors.
Collapse
Affiliation(s)
- Siu Woon Cheng
- Department of Otorhinolaryngology, Head and Neck Surgery, Tuen Mun Hospital, 23 Tsing Chung Koon Road, Tuen Mun, New Territories, Hong Kong.
| | - Kwok Hung Vincent Leung
- Department of Otorhinolaryngology, Head and Neck Surgery, Tuen Mun Hospital, 23 Tsing Chung Koon Road, Tuen Mun, New Territories, Hong Kong
| | - Kar Cheong Jason Mok
- Department of Otorhinolaryngology, Head and Neck Surgery, Tuen Mun Hospital, 23 Tsing Chung Koon Road, Tuen Mun, New Territories, Hong Kong
| | - Kong Wah Yeung
- Department of Otorhinolaryngology, Head and Neck Surgery, Tuen Mun Hospital, 23 Tsing Chung Koon Road, Tuen Mun, New Territories, Hong Kong
| | - Sin Yee Ivy Wong
- Department of Speech Therapy, Tuen Mun Hospital, Tuen Mun, Hong Kong
| | - Yin Ling Lam
- Department of Speech Therapy, Tuen Mun Hospital, Tuen Mun, Hong Kong
| | - Ka Man Ip
- Department of Speech Therapy, Tuen Mun Hospital, Tuen Mun, Hong Kong
| | - Yin Wing Lok
- Department of Speech Therapy, Tuen Mun Hospital, Tuen Mun, Hong Kong
| | - Andrew Chun Lok Wong
- Department of Otorhinolaryngology, Head and Neck Surgery, Tuen Mun Hospital, 23 Tsing Chung Koon Road, Tuen Mun, New Territories, Hong Kong
| |
Collapse
|
23
|
Pattani N, Sanghera J, Langridge BJ, Frommer ML, Abu-Hanna J, Butler P. Exploring the mechanisms behind autologous lipotransfer for radiation-induced fibrosis: A systematic review. PLoS One 2024; 19:e0292013. [PMID: 38271326 PMCID: PMC10810439 DOI: 10.1371/journal.pone.0292013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/11/2023] [Indexed: 01/27/2024] Open
Abstract
AIM Radiation-induced fibrosis is a recognised consequence of radiotherapy, especially after multiple and prolonged dosing regimens. There is no definitive treatment for late-stage radiation-induced fibrosis, although the use of autologous fat transfer has shown promise. However, the exact mechanisms by which this improves radiation-induced fibrosis remain poorly understood. We aim to explore existing literature on the effects of autologous fat transfer on both in-vitro and in-vivo radiation-induced fibrosis models, and to collate potential mechanisms of action. METHOD PubMed, Cochrane reviews and Scopus electronic databases from inception to May 2023 were searched. Our search strategy combined both free-text terms with Boolean operators, derived from synonyms of adipose tissue and radiation-induced fibrosis. RESULTS The search strategy produced 2909 articles. Of these, 90 underwent full-text review for eligibility, yielding 31 for final analysis. Nine conducted in-vitro experiments utilising a co-culture model, whilst 25 conducted in-vivo experiments. Interventions under autologous fat transfer included adipose-derived stem cells, stromal vascular function, whole fat and microfat. Notable findings include downregulation of fibroblast proliferation, collagen deposition, epithelial cell apoptosis, and proinflammatory processes. Autologous fat transfer suppressed hypoxia and pro-inflammatory interferon-γ signalling pathways, and tissue treated with adipose-derived stem cells stained strongly for anti-inflammatory M2 macrophages. Although largely proangiogenic initially, studies show varying effects on vascularisation. There is early evidence that adipose-derived stem cell subgroups may have different functional properties. CONCLUSION Autologous fat transfer functions through pro-angiogenic, anti-fibrotic, immunomodulatory, and extracellular matrix remodelling properties. By characterising these mechanisms, relevant drug targets can be identified and used to further improve clinical outcomes in radiation-induced fibrosis. Further research should focus on adipose-derived stem cell sub-populations and augmentation techniques such as cell-assisted lipotransfer.
Collapse
Affiliation(s)
| | | | - Benjamin J. Langridge
- Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
| | - Marvin L. Frommer
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
| | - Jeries Abu-Hanna
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
- Division of Medical Sciences, University of Oxford, Oxford, United Kingdom
| | - Peter Butler
- Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
24
|
Malekzadeh H, Surucu Y, Chinnapaka S, Yang KS, Arellano JA, Samadi Y, Epperly MW, Greenberger JS, Rubin JP, Ejaz A. Metformin and adipose-derived stem cell combination therapy alleviates radiation-induced skin fibrosis in mice. Stem Cell Res Ther 2024; 15:13. [PMID: 38185658 PMCID: PMC10773046 DOI: 10.1186/s13287-023-03627-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/26/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Radiation therapy often leads to late radiation-induced skin fibrosis (RISF), causing movement impairment and discomfort. We conducted a comprehensive study to assess the effectiveness of metformin and adipose-derived stem cells (ASCs), whether autologous or allogeneic, individually or in combination therapy, in mitigating RISF. METHODS Using a female C57BL/6J mouse model subjected to hind limb irradiation as a representative RISF model, we evaluated metformin, ASCs, or their combination in two contexts: prophylactic (started on day 1 post-irradiation) and therapeutic (initiated on day 14 post-irradiation, coinciding with fibrosis symptoms). We measured limb movement, examined skin histology, and analyzed gene expression to assess treatment efficacy. RESULTS Prophylactic metformin and ASCs, whether autologous or allogeneic, effectively prevented late fibrosis, with metformin showing promising results. However, combination therapy did not provide additional benefits when used prophylactically. Autologous ASCs, alone or with metformin, proved most effective against late-stage RISF. Prophylactic intervention outperformed late therapy for mitigating radiation skin damage. Co-culture studies revealed that ASCs and metformin downregulated inflammation and fibrotic gene expression in both mouse and human fibroblasts. CONCLUSIONS Our study suggests metformin's potential as a prophylactic measure to prevent RISF, and the combination of ASCs and metformin holds promise for late-stage RISF treatment. These findings have clinical implications for improving the quality of life for those affected by radiation-induced skin fibrosis.
Collapse
Affiliation(s)
- Hamid Malekzadeh
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
| | - Yusuf Surucu
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
| | - Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
| | - Katherine S Yang
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
| | - José A Arellano
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
| | - Yasamin Samadi
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA
- McGowan Institute, University of Pittsburgh, Pittsburgh, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh Medical Center, 3550 Terrace Street, 6B Scaife Hall, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
25
|
Sachdev S. A friend in a forest of radiation-immune interactions: BAMBI improves antitumor effects by limiting radioresistance. J Clin Invest 2023; 133:e176061. [PMID: 38099499 PMCID: PMC10721139 DOI: 10.1172/jci176061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Radiation therapy (RT) remains one of the most effective and utilized oncologic treatments available. While it can directly yield tumor cell death, its impact on the immune microenvironment is more complex, promoting either an antitumor response or, conversely, a tumor-promoting state. TGF-β, induced by RT, yields a more immunosuppressive environment, including potentially blunting response to immune-checkpoint blockade. In this issue of the JCI, Wang and colleagues demonstrate that RT reduced expression of bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI), a TGF-β pseudoreceptor. Limiting this effect, or increasing BAMBI, improved RT-induced tumor cell killing, tumor response, and antitumor immune effects. This realization points to a pathway of potential clinical translation.
Collapse
|
26
|
McMahon RA, D'Souza C, Neeson PJ, Siva S. Innate immunity: Looking beyond T-cells in radiation and immunotherapy combinations. Neoplasia 2023; 46:100940. [PMID: 37913654 PMCID: PMC10637988 DOI: 10.1016/j.neo.2023.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Radiation therapy is an established and effective anti-cancer treatment modality. Extensive pre-clinical experimentation has demonstrated that the pro-inflammatory properties of irradiation may be synergistic with checkpoint immunotherapy. Radiation induces double-stranded DNA breaks (dsDNA). Sensing of the dsDNA activates the cGAS/STING pathway, producing Type 1 interferons essential to recruiting antigen-presenting cells (APCs). Radiation promotes cytotoxic CD8 T-cell recruitment by releasing tumour-associated antigens captured and cross-presented by surveying antigen-presenting cells. Radiation-induced vascular normalisation may further promote T-cell trafficking and drug delivery. Radiation is also immunosuppressive. Recruitment of regulatory T cells (Tregs) and innate cells such as myeloid-derived suppressive cells (m-MDSCs) all counteract the immunostimulatory properties of radiation. Many innate immune cell types operate at the interface of the adaptive immune response. Innate immune cells, such as m-MDSCs, can exert their immunosuppressive effects by expressing immune checkpoints such as PD-L1, further highlighting the potential of combined radiation and checkpoint immunotherapy. Several early-phase clinical studies investigating the combination of radiation and immunotherapy have been disappointing. A greater appreciation of radiotherapy's impact on the innate immune system is essential to optimise radioimmunotherapy combinations. This review will summarise the impact of radiotherapy on crucial cells of the innate immune system and vital immunosuppressive cytokines.
Collapse
Affiliation(s)
- R A McMahon
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.
| | - C D'Souza
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - P J Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - S Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Atajanova T, Rahman MM, Konieczkowski DJ, Morris ZS. Radiation-associated secondary malignancies: a novel opportunity for applying immunotherapies. Cancer Immunol Immunother 2023; 72:3445-3452. [PMID: 37658906 PMCID: PMC10992240 DOI: 10.1007/s00262-023-03532-1] [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] [Received: 04/17/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Radiation is commonly used as a treatment intended to cure or palliate cancer patients. Despite remarkable advances in the precision of radiotherapy delivery, even the most advanced forms inevitably expose some healthy tissues surrounding the target site to radiation. On rare occasions, this results in the development of radiation-associated secondary malignancies (RASM). RASM are typically high-grade and carry a poorer prognosis than their non-radiated counterparts. RASM are characterized by a high mutation burden, increased T cell infiltration, and a microenvironment that bears unique inflammatory signatures of prior radiation, including increased expression of various cytokines (e.g., TGF-β, TNF-α, IL4, and IL10). Interestingly, these cytokines have been shown to up-regulate the expression of PD-1 and/or PD-L1-an immune checkpoint receptor/ligand pair that is commonly targeted by immune checkpoint blocking immunotherapies. Here, we review the current understanding of the tumor-immune interactions in RASM, highlight the distinct clinical and molecular characteristics of RASM that may render them immunologically "hot," and propose a rationale for the formal testing of immune checkpoint blockade as a treatment approach for patients with RASM.
Collapse
Affiliation(s)
- Tavus Atajanova
- Biochemistry and Biophysics Program, Amherst College, Amherst, MA, 01002, USA
- Department of Sociology, Amherst College, Amherst, MA, 01002, USA
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53726, USA
| | - Md Mahfuzur Rahman
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53726, USA
| | - David J Konieczkowski
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53726, USA.
| |
Collapse
|
28
|
Kasikasetsiri J, Apaijai N, Aschaitrakool Y, Kerdphoo S, Sriyaranya N, Chattipakorn N, Chattipakorn SC. Hyperbaric oxygen therapy restores wound healing in irradiated gingiva to a similar level to that in healthy gingiva. J Wound Care 2023; 32:676-684. [PMID: 37830829 DOI: 10.12968/jowc.2023.32.10.676] [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: 10/14/2023]
Abstract
OBJECTIVE This study aimed to investigate the involvement of mitochondrial biogenesis, and determine the extent of fibroblast proliferation and cellular apoptosis, in the gingiva of patients who had undergone head and neck radiation, after receiving hyperbaric oxygen therapy (HBOT), in comparison with normal gingiva. METHOD A total of 16 patients who had undergone head and neck radiation with HBOT and six healthy subjects were included in the study. After the completion of radiation therapy, patients received HBOT at 2 ATA for 90 minutes per session, and for 20 sessions per patient. Samples of gingival tissues were then taken. The levels of: transforming growth factor beta (TGF-β); phospho-nuclear factor kappa-light-chain-enhancer of activated B cells (p-NFϰB); nuclear factor kappa-light-chain-enhancer of activated B cells (NFϰB); proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α); phospho-dynamin-related protein 1 at ser616 (p-Drp1ser616); dynamin-related protein 1 (Drp1); Bcl-2-associated X-protein (Bax); and B-cell lymphoma 2 (Bcl-2) were determined using a Western blot. Independent t-test and Chi-squared tests were used in the study. RESULTS There were no differences in the levels of TGF-β, p-NFϰB, NFϰB, p-Drp1ser616, Drp1, Bax and Bcl-2 between the two groups. However, the level of PGC-1α was greater in irradiated gingival tissues with HBOT than in the healthy gingiva. CONCLUSION Radiation-induced impaired wound healing can be improved by HBOT as indicated by levels of apoptosis, mitochondrial dynamics, cell proliferation and inflammation in irradiated gingiva with HBOT to a similar level to normal healthy gingiva. These findings may occur through an increase in mitochondrial biogenesis following HBOT.
Collapse
Affiliation(s)
- Juthathip Kasikasetsiri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Yuthakran Aschaitrakool
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nutchada Sriyaranya
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
29
|
Brooks KA, Gross JH. Radiotherapy-induced Pathology of the Ear. Otolaryngol Clin North Am 2023; 56:977-985. [PMID: 37414656 DOI: 10.1016/j.otc.2023.05.013] [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: 07/08/2023]
Abstract
Acute radiotherapy (RT)-induced external ear soft tissue changes start with erythema and dry desquamation and may progress to moist desquamation and epidermal ulceration. Chronic RT-induced changes include epithelial atrophy and subcutaneous fibrosis. Although RT-induced radiation dermatitis has been well studied, interventions for soft tissue disease involving the external auditory canal (EAC) warrant investigation. Medical management includes topical steroid treatment for EAC radiation dermatitis and topical antibiotic therapy for suppurative otitis externa. Hyperbaric oxygen and pentoxifylline-vitamin E therapy have shown promise for other applications, but their clinical effect on soft tissue EAC disease is currently undefined.
Collapse
Affiliation(s)
- Kaitlyn A Brooks
- Department of Otolaryngology-Head and Neck Surgery, Emory University, Atlanta, GA 30308, USA
| | - Jennifer H Gross
- Department of Otolaryngology-Head and Neck Surgery, Emory University, Atlanta, GA 30308, USA.
| |
Collapse
|
30
|
Chinnapaka S, Yang KS, Surucu Y, Bengur FB, Arellano JA, Tirmizi Z, Malekzadeh H, Epperly MW, Hou W, Greenberger JS, Rubin JP, Ejaz A. Human adipose ECM alleviates radiation-induced skin fibrosis via endothelial cell-mediated M2 macrophage polarization. iScience 2023; 26:107660. [PMID: 37705953 PMCID: PMC10495661 DOI: 10.1016/j.isci.2023.107660] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/30/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Radiation therapy can lead to late radiation-induced skin fibrosis (RISF), causing movement restriction, pain, and organ dysfunction. This study evaluated adipose-derived extracellular matrix (Ad-ECM) as a mitigator of RISF. Female C57BL/6J mice that were irradiated developed fibrosis, which was mitigated by a single local Ad-ECM injection, improving limb movement and reducing epithelium thickness and collagen deposition. Ad-ECM treatment resulted in decreased expression of pro-inflammatory and fibrotic genes, and upregulation of anti-inflammatory cytokines, promoting M2 macrophage polarization. Co-culture of irradiated human fibroblasts with Ad-ECM down-modulated fibrotic gene expression and enhanced bone marrow cell migration. Ad-ECM treatment also increased interleukin (IL)-4, IL-5, and IL-15 expression in endothelial cells, stimulating M2 macrophage polarization and alleviating RISF. Prophylactic use of Ad-ECM showed effectiveness in mitigation. This study suggests Ad-ECM's potential in treating chronic-stage fibrosis.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine S. Yang
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yusuf Surucu
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fuat B. Bengur
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - José A. Arellano
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zayaan Tirmizi
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hamid Malekzadeh
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Wen Hou
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - J. Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
31
|
King SN, Kaissieh N, Haxton C, Shojaei M, Malott L, Devara L, Thompson R, Osman KL, Millward J, Blackburn M, Lever TE. Radiation induced changes in profibrotic markers in the submental muscles and their correlation with tongue movement. PLoS One 2023; 18:e0287044. [PMID: 37352202 PMCID: PMC10289304 DOI: 10.1371/journal.pone.0287044] [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: 11/04/2022] [Accepted: 05/29/2023] [Indexed: 06/25/2023] Open
Abstract
Swallowing impairment is a major complication of radiation treatment for oropharyngeal cancers. Developing targeted therapies that improve swallowing outcomes relies on an understanding of the mechanisms that influence motor function after radiation treatment. The purpose of this study was to determine whether there is a correlation between radiation induced changes in tongue movement and structural changes in irradiated submental muscles, as well as assess other possible causes for dysfunction. We hypothesized that a clinically relevant total radiation dose to the submental muscles would result in: a) quantifiable changes in tongue strength and displacement during drinking two months post treatment; and b) a profibrotic response and/or fiber type transition in the irradiated tissue. Sprague-Dawley adult male rats received radiation to the submental muscles at total dose-volumes known to provoke dysphagia in humans. A clinical linear accelerator administered 8 fractions of 8Gy for a total of 64Gy. Comparisons were made to sham-treated rats that received anesthesia only. Swallowing function was assessed using videofluoroscopy and tongue strength was analyzed via force lickometer. TGFβ1 expression was analyzed via ELISA. The amount of total collagen was analyzed by picrosirius red staining. Immunofluorescence was used to assess fiber type composition and size. Significant changes in licking function during drinking were observed at two months post treatment, including a slower lick rate and reduced tongue protrusion during licking. In the mylohyoid muscle, significant increases in TGFβ1 protein expression were found post radiation. Significant increases in the percentage of collagen content were observed in the irradiated geniohyoid muscle. No changes in fiber type expression were observed. Results indicate a profibrotic transition within the irradiated swallowing muscles that contributes to tongue dysfunction post-radiation treatment.
Collapse
Affiliation(s)
- Suzanne N. King
- Department of Otolaryngology–Head and Neck Surgery and Communicative Disorders, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Nada Kaissieh
- Department of Otolaryngology–Head and Neck Surgery and Communicative Disorders, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Chandler Haxton
- Department of Otolaryngology—Head and Neck Surgery, University of Missouri School of Medicine, Columbia, MO, United States of America
| | - Marjan Shojaei
- Department of Radiation Oncology, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Luke Malott
- Department of Otolaryngology–Head and Neck Surgery and Communicative Disorders, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Lekha Devara
- Department of Otolaryngology–Head and Neck Surgery and Communicative Disorders, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Rebecca Thompson
- Department of Otolaryngology—Head and Neck Surgery, University of Missouri School of Medicine, Columbia, MO, United States of America
| | - Kate L. Osman
- Department of Otolaryngology—Head and Neck Surgery, University of Missouri School of Medicine, Columbia, MO, United States of America
| | - Jessica Millward
- Department of Otolaryngology–Head and Neck Surgery and Communicative Disorders, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Megan Blackburn
- Department of Radiation Oncology, School of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Teresa E. Lever
- Department of Otolaryngology—Head and Neck Surgery, University of Missouri School of Medicine, Columbia, MO, United States of America
| |
Collapse
|
32
|
Prescher H, Froimson JR, Hanson SE. Deconstructing Fat to Reverse Radiation Induced Soft Tissue Fibrosis. Bioengineering (Basel) 2023; 10:742. [PMID: 37370673 PMCID: PMC10295516 DOI: 10.3390/bioengineering10060742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Adipose tissue is composed of a collection of cells with valuable structural and regenerative function. Taken as an autologous graft, these cells can be used to address soft tissue defects and irregularities, while also providing a reparative effect on the surrounding tissues. Adipose-derived stem or stromal cells are primarily responsible for this regenerative effect through direct differentiation into native cells and via secretion of numerous growth factors and cytokines that stimulate angiogenesis and disrupt pro-inflammatory pathways. Separating adipose tissue into its component parts, i.e., cells, scaffolds and proteins, has provided new regenerative therapies for skin and soft tissue pathology, including that resulting from radiation. Recent studies in both animal models and clinical trials have demonstrated the ability of autologous fat grafting to reverse radiation induced skin fibrosis. An improved understanding of the complex pathologic mechanism of RIF has allowed researchers to harness the specific function of the ASCs to engineer enriched fat graft constructs to improve the therapeutic effect of AFG.
Collapse
Affiliation(s)
| | | | - Summer E. Hanson
- Section of Plastic & Reconstructive Surgery, University of Chicago Medical Center, Chicago, IL 60615, USA
| |
Collapse
|
33
|
Sterling J, Rahman SN, Varghese A, Angulo JC, Nikolavsky D. Complications after Prostate Cancer Treatment: Pathophysiology and Repair of Post-Radiation Urethral Stricture Disease. J Clin Med 2023; 12:3950. [PMID: 37373644 DOI: 10.3390/jcm12123950] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Radiation therapy (RT) in the management of pelvic cancers remains a clinical challenge to urologists given the sequelae of urethral stricture disease secondary to fibrosis and vascular insults. The objective of this review is to understand the physiology of radiation-induced stricture disease and to educate urologists in clinical practice regarding future prospective options clinicians have to deal with this condition. The management of post-radiation urethral stricture consists of conservative, endoscopic, and primary reconstructive options. Endoscopic approaches remain an option, but with limited long-term success. Despite concerns with graft take, reconstructive options such as urethroplasties in this population with buccal grafts have shown long-term success rates ranging from 70 to 100%. Robotic reconstruction is augmenting previous options with faster recovery times. Radiation-induced stricture disease is challenging with multiple interventions available, but with successful outcomes demonstrated in various cohorts including urethroplasties with buccal grafts and robotic reconstruction.
Collapse
Affiliation(s)
- Joshua Sterling
- Yale School of Medicine, 20 York Street, New Haven, CT 06511, USA
| | - Syed N Rahman
- Yale School of Medicine, 20 York Street, New Haven, CT 06511, USA
| | - Ajin Varghese
- New York College of Osteopathic Medicine, 8000 Old Westbury, Glen Head, NY 11545, USA
| | - Javier C Angulo
- Faculty of Biomedical Sciences, Universidad Europea, 28905 Madrid, Spain
| | | |
Collapse
|
34
|
Ng JPZ, Lam WYH, Pow EHN, Botelho MG. A qualitative analysis of patient's lived experience on their treatment journey with nasopharyngeal carcinoma. J Dent 2023; 134:104518. [PMID: 37088259 DOI: 10.1016/j.jdent.2023.104518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/25/2023] Open
Abstract
OBJECTIVE To explore and analyse the perspective of patients undergoing and recovering from nasopharyngeal carcinoma (NPC) therapy. METHODS Thirty-three NPC patients at different stages of treatment were enrolled. Seven were actively undergoing treatment, 13 were immediately post-treatment, and 13 were long-term. Patients were interviewed using a structured questionnaire based on a review of the literature that covered different phases of their treatment journey. The interview was recorded and transcribed for qualitative data analysis using a thematic inductive-deductive approach. RESULTS Three main domains embracing aspects of NPC patients' experiences were identified; side effects, psychosocial well-being, and the role and support of healthcare workers. Side effects were experienced orally, locally, and systemically. Oral side effects (oral mucositis, xerostomia, altered taste, dysphagia) were the most significant challenge experienced by NPC patients. Locally, skin injury (desquamation, fibrosis, darkening of the skin, erythema, pruritus, and swelling around the neck region) and hair loss, resolved after cessation of therapy. Systemic side effects from the treatment were related to general weakness, weight loss and nausea. The psychosocial well-being of NPC patients was influenced by a range of issues including support (healthcare workers and family), pain management, functional limitations, nutritional needs, perceived level of information, emotion, and finances. CONCLUSION NPC patients were significantly impacted based on the diagnosis, treatment and recovery phase affecting them locally, systemically, and psychologically. The role of family and healthcare staff was also influential in the overall treatment experience, and they have key roles to play in facilitating patients along their treatment journey. CLINICAL SIGNIFICANCE Oral and general side effects from NPC treatment have significant impact on patients physical and emotional well-being. It is important for healthcare providers to have insights of these so as to understand and support patients during their treatment journey and recovery and be able to empathetically facilitate their clinical management.
Collapse
Affiliation(s)
- Joanne Pui Zhee Ng
- Graduate Student in Prosthodontic, Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Walter Yu Hang Lam
- Clinical Assistant Professor in Prosthodontics, Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Edmond Ho Nang Pow
- Clinical Associate Professor in Prosthodontics, Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Michael G Botelho
- Clinical Professor in Prosthodontics, Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
35
|
Topkan E, Kucuk A, Somay E, Yilmaz B, Pehlivan B, Selek U. Review of Osteoradionecrosis of the Jaw: Radiotherapy Modality, Technique, and Dose as Risk Factors. J Clin Med 2023; 12:3025. [PMID: 37109361 PMCID: PMC10143049 DOI: 10.3390/jcm12083025] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Radiotherapy (RT) or concurrent chemoradiotherapy (CCRT) is the cornerstone of organ-sparing or adjuvant therapy for nearly all head and neck cancers. Unfortunately, aggressive RT or CCRT can result in severe late toxicities, such as osteoradionecrosis of the jaws (ORNJ). The incidence of ORNJ is currently less than 5-6% due to advances in dental preventive care programs, RT planning systems, and RT techniques. Although numerous patient-, tumor-, and treatment-related factors may influence the incidence rates of ORNJ, RT modality (equipment), technique, and dose-volume-related factors are three of the most influential factors. This is mainly because different RT equipment and techniques have different levels of success at delivering the prescribed dose to the focal volume of the treatment while keeping the "organ at risk" safe. ORNJ risk is ultimately determined by mandibular dose, despite the RT technique and method being known predictors. Regardless of the photon delivery method, the radiobiological effects will be identical if the total dose, dose per fraction, and dose distribution within the tissue remain constant. Therefore, contemporary RT procedures mitigate this risk by reducing mandibular dosages rather than altering the ionizing radiation behavior in irradiated tissues. In light of the paucity of studies that have examined the impact of RT modality, technique, and dose-volume-related parameters, as well as their radiobiological bases, the present review aims to provide a comprehensive overview of the published literature on these specific issues to establish a common language among related disciplines and provide a more reliable comparison of research results.
Collapse
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Medical Faculty, Baskent University, Adana 01120, Turkey
| | - Ahmet Kucuk
- Clinics of Radiation Oncology, Mersin City Education and Research Hospital, Mersin 33160, Turkey
| | - Efsun Somay
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Baskent University, Ankara 06490, Turkey
| | - Busra Yilmaz
- Department of Oral and Maxillofacial Radiology, Faculty of Dentistry, Baskent University, Ankara 06490, Turkey
| | - Berrin Pehlivan
- Department of Radiation Oncology, Bahcesehir University, Istanbul 34349, Turkey
| | - Ugur Selek
- Department of Radiation Oncology, School of Medicine, Koc University, Istanbul 34450, Turkey
- Department of Radiation Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| |
Collapse
|
36
|
Hanson I, Pitman KE, Edin NFJ. The Role of TGF-β3 in Radiation Response. Int J Mol Sci 2023; 24:ijms24087614. [PMID: 37108775 PMCID: PMC10141893 DOI: 10.3390/ijms24087614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Transforming growth factor-beta 3 (TGF-β3) is a ubiquitously expressed multifunctional cytokine involved in a range of physiological and pathological conditions, including embryogenesis, cell cycle regulation, immunoregulation, and fibrogenesis. The cytotoxic effects of ionizing radiation are employed in cancer radiotherapy, but its actions also influence cellular signaling pathways, including that of TGF-β3. Furthermore, the cell cycle regulating and anti-fibrotic effects of TGF-β3 have identified it as a potential mitigator of radiation- and chemotherapy-induced toxicity in healthy tissue. This review discusses the radiobiology of TGF-β3, its induction in tissue by ionizing radiation, and its potential radioprotective and anti-fibrotic effects.
Collapse
Affiliation(s)
- Ingunn Hanson
- Department of Physics, University of Oslo, 0371 Oslo, Norway
| | | | - Nina F J Edin
- Department of Physics, University of Oslo, 0371 Oslo, Norway
| |
Collapse
|
37
|
Abstract
Head and neck cancer (HNC) survivorship is increasing, and with it, a shift in treatment practices has occurred. Radical surgical resections for the treatment of HNC have decreased, and organ preservation treatments have increased. Although effective in treating HNC, chemoradiation therapy toxicities can be detrimental to a patient's overall health, nutrition status, and quality of life (QOL). Considering that dysphagia is typically a driving element of dysfunction, speech-language pathologists are vital to the prehabilitation phase. Prehabilitation programs include a variety of components, with the primary goal being to improve functional and QOL outcomes posttreatment.
Collapse
Affiliation(s)
- Mary Caroline Murray
- University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA.
| | - Anne Kane
- University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
38
|
Callaghan CM, Abukhiran IM, Masaadeh A, Van Rheeden RV, Kalen AL, Rodman SN, Petronek MS, Mapuskar KA, George BN, Coleman MC, Goswami PC, Allen BG, Spitz DR, Caster JM. Manipulation of Redox Metabolism Using Pharmacologic Ascorbate Opens a Therapeutic Window for Radio-Sensitization by ATM Inhibitors in Colorectal Cancer. Int J Radiat Oncol Biol Phys 2023; 115:933-944. [PMID: 36228747 PMCID: PMC9974877 DOI: 10.1016/j.ijrobp.2022.10.012] [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: 08/15/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Ataxia telangiectasia mutated kinase (ATM) inhibitors are potent radiosensitizers that regulate DNA damage responses and redox metabolism, but they have not been translated clinically because of the potential for excess normal tissue toxicity. Pharmacologic ascorbate (P-AscH-; intravenous administration achieving mM plasma concentrations) selectively enhances H2O2-induced oxidative stress and radiosensitization in tumors while acting as an antioxidant and mitigating radiation damage in normal tissues including the bowel. We hypothesized that P-AscH- could enhance the therapeutic index of ATM inhibitor-based chemoradiation by simultaneously enhancing the intended effects of ATM inhibitors in tumors and mitigating off-target effects in adjacent normal tissues. METHODS AND MATERIALS Clonogenic survival was assessed in human (human colon tumor [HCT]116, SW480, HT29) and murine (CT26, MC38) colorectal tumor lines and normal cells (human umbilical vein endothelial cell, FHs74) after radiation ± DNA repair inhibitors ± P-AscH-. Tumor growth delay was assessed in mice with HCT116 or MC38 tumors after fractionated radiation (5 Gy × 3) ± the ATM inhibitor KU60019 ± P-AscH-. Intestinal injury, oxidative damage, and transforming growth factor β immunoreactivity were quantified using immunohistochemistry after whole abdominal radiation (10 Gy) ± KU60019 ± P-AscH-. Cell cycle distribution and ATM subcellular localization were assessed using flow cytometry and immunohistochemistry. The role of intracellular H2O2 fluxes was assessed using a stably expressed doxycycline-inducible catalase transgene. RESULTS KU60019 with P-AscH- enhanced radiosensitization in colorectal cancer models in vitro and in vivo by H2O2-dependent oxidative damage to proteins and enhanced DNA damage, abrogation of the postradiation G2 cell cycle checkpoint, and inhibition of ATM nuclear localization. In contrast, concurrent P-AscH- markedly reduced intestinal toxicity and oxidative damage with KU60019. CONCLUSIONS We provide evidence that redox modulating drugs, such as P-AscH-, may facilitate the clinical translation of ATM inhibitors by enhancing tumor radiosensitization while simultaneously protecting normal tissues.
Collapse
Affiliation(s)
- Cameron M Callaghan
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Ibrahim M Abukhiran
- Department of Pathology, University of Iowa Hospitals and Clinics and Carver College of Medicine, Iowa City, Iowa
| | - Amr Masaadeh
- Department of Pathology, University of Iowa Hospitals and Clinics and Carver College of Medicine, Iowa City, Iowa
| | | | - Amanda L Kalen
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Samuel N Rodman
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Michael S Petronek
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Kranti A Mapuskar
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Benjamin N George
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Mitchell C Coleman
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Prabhat C Goswami
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Bryan G Allen
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Douglas R Spitz
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Joseph M Caster
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
39
|
Tramm T, Kaidar-Person O. Optimising post-operative radiation therapy after oncoplastic and reconstructive procedures. Breast 2023; 69:366-374. [PMID: 37023565 PMCID: PMC10119683 DOI: 10.1016/j.breast.2023.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/16/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023] Open
Abstract
Surgical techniques for breast cancer have been refined over the past decades to deliver an aesthetic outcome as close as possible to the contralateral intact breast. Current surgery further allows excellent aesthetic outcome even in case of mastectomy, by performing skin sparing or nipple sparing mastectomy in combination with breast reconstruction. In this review we discuss how to optimise post-operative radiation therapy after oncoplastic and breast reconstructive procedures, including dose, fractionation, volumes, surgical margins, and boost application.
Collapse
|
40
|
He L, Yu X, Li W. Recent Progress and Trends in X-ray-Induced Photodynamic Therapy with Low Radiation Doses. ACS NANO 2022; 16:19691-19721. [PMID: 36378555 DOI: 10.1021/acsnano.2c07286] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The prominence of photodynamic therapy (PDT) in treating superficial skin cancer inspires innovative solutions for its congenitally deficient shadow penetration of the visible-light excitation. X-ray-induced photodynamic therapy (X-PDT) has been proven to be a successful technique in reforming the conventional PDT for deep-seated tumors by creatively utilizing penetrating X-rays as external excitation sources and has witnessed rapid developments over the past several years. Beyond the proof-of-concept demonstration, recent advances in X-PDT have exhibited a trend of minimizing X-ray radiation doses to quite low values. As such, scintillating materials used to bridge X-rays and photosensitizers play a significant role, as do diverse well-designed irradiation modes and smart strategies for improving the tumor microenvironment. Here in this review, we provide a comprehensive summary of recent achievements in X-PDT and highlight trending efforts using low doses of X-ray radiation. We first describe the concept of X-PDT and its relationships with radiodynamic therapy and radiotherapy and then dissect the mechanism of X-ray absorption and conversion by scintillating materials, reactive oxygen species evaluation for X-PDT, and radiation side effects and clinical concerns on X-ray radiation. Finally, we discuss a detailed overview of recent progress regarding low-dose X-PDT and present perspectives on possible clinical translation. It is expected that the pursuit of low-dose X-PDT will facilitate significant breakthroughs, both fundamentally and clinically, for effective deep-seated cancer treatment in the near future.
Collapse
|
41
|
The Normal, the Radiosensitive, and the Ataxic in the Era of Precision Radiotherapy: A Narrative Review. Cancers (Basel) 2022; 14:cancers14246252. [PMID: 36551737 PMCID: PMC9776433 DOI: 10.3390/cancers14246252] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: radiotherapy is a cornerstone of cancer treatment. When delivering a tumoricidal dose, the risk of severe late toxicities is usually kept below 5% using dose-volume constraints. However, individual radiation sensitivity (iRS) is responsible (with other technical factors) for unexpected toxicities after exposure to a dose that induces no toxicity in the general population. Diagnosing iRS before radiotherapy could avoid unnecessary toxicities in patients with a grossly normal phenotype. Thus, we reviewed iRS diagnostic data and their impact on decision-making processes and the RT workflow; (2) Methods: following a description of radiation toxicities, we conducted a critical review of the current state of the knowledge on individual determinants of cellular/tissue radiation; (3) Results: tremendous advances in technology now allow minimally-invasive genomic, epigenetic and functional testing and a better understanding of iRS. Ongoing large translational studies implement various tests and enriched NTCP models designed to improve the prediction of toxicities. iRS testing could better support informed radiotherapy decisions for individuals with a normal phenotype who experience unusual toxicities. Ethics of medical decisions with an accurate prediction of personalized radiotherapy's risk/benefits and its health economics impact are at stake; (4) Conclusions: iRS testing represents a critical unmet need to design personalized radiotherapy protocols relying on extended NTCP models integrating iRS.
Collapse
|
42
|
A role for endothelial alpha-mannosidase MAN1C1 in radiation-induced immune cell recruitment. iScience 2022; 25:105482. [DOI: 10.1016/j.isci.2022.105482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/06/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
|
43
|
Buntinx F, Lebeau A, Gillot L, Baudin L, Ndong Penda R, Morfoisse F, Lallemand F, Vottero G, Nizet C, Nizet JL, Blacher S, Noel A. Single and combined impacts of irradiation and surgery on lymphatic vasculature and fibrosis associated to secondary lymphedema. Front Pharmacol 2022; 13:1016138. [PMID: 36330083 PMCID: PMC9622766 DOI: 10.3389/fphar.2022.1016138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Lymphedema (LD) refers to a condition of lymphatic dysfunction associated with excessive fluid accumulation, fibroadipose tissue deposition and swelling. In industrialized countries, LD development mainly results from a local disruption of the lymphatic network by an infection or cancer-related surgery (secondary LD). In the absence of efficient therapy, animal models are needed to decipher the cellular and molecular mechanisms underlying LD and test putative drugs. In this study, we optimized and characterized a murine model of LD that combines an irradiation of the mice hind limb and a radical surgery (lymph node resection associated to lymphatic vessel ligation). We investigated the respective roles of irradiation and surgery in LD formation by comparing their impacts, alone or in combination (with different intervention sequences), on eight different features of the pathology: swelling (paw thickness), indocyanine green (ICG) clearance, lymphatic vasculature remodeling, epidermal and dermal thickening, adipocyte accumulation, inflammatory cell infiltration and collagen deposition. This study supports the importance of radiation prior to surgery to experimentally induce a rapid, severe and sustained tissue remodeling harboring the different hallmarks of LD. We provide the first experimental evidence for an excessive deposition of periostin (POSTN) and tenascin-C (TNC) in LD. Through a computerized method of digital image quantification, we established the spatial map of lymphatic expansion, as well as collagen, POSTN and TNC deposition in papillary and reticular dermis of lymphedematous skins. This mouse model is available to study the patho-physiology of LD and test potential therapeutic targets.
Collapse
Affiliation(s)
- F. Buntinx
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - A. Lebeau
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - L. Gillot
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - L. Baudin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - R. Ndong Penda
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - F. Morfoisse
- U1297-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Toulouse, Toulouse, France
| | - F. Lallemand
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
- Department of Radiotherapy-Oncology, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - G. Vottero
- Department of Plastic and Reconstructive Surgery, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - C. Nizet
- Department of Plastic and Reconstructive Surgery, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - J. L. Nizet
- Department of Plastic and Reconstructive Surgery, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Liège, Belgium
| | - S. Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
| | - A. Noel
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège (ULiège), Sart-Tilman, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
- *Correspondence: A. Noel,
| |
Collapse
|
44
|
Potential Molecular Mechanisms behind the Ultra-High Dose Rate "FLASH" Effect. Int J Mol Sci 2022; 23:ijms232012109. [PMID: 36292961 PMCID: PMC9602825 DOI: 10.3390/ijms232012109] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/26/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
FLASH radiotherapy, or the delivery of a dose at an ultra-high dose rate (>40 Gy/s), has recently emerged as a promising tool to enhance the therapeutic index in cancer treatment. The remarkable sparing of normal tissues and equivalent tumor control by FLASH irradiation compared to conventional dose rate irradiation—the FLASH effect—has already been demonstrated in several preclinical models and even in a first patient with T-cell cutaneous lymphoma. However, the biological mechanisms responsible for the differential effect produced by FLASH irradiation in normal and cancer cells remain to be elucidated. This is of great importance because a good understanding of the underlying radiobiological mechanisms and characterization of the specific beam parameters is required for a successful clinical translation of FLASH radiotherapy. In this review, we summarize the FLASH investigations performed so far and critically evaluate the current hypotheses explaining the FLASH effect, including oxygen depletion, the production of reactive oxygen species, and an altered immune response. We also propose a new theory that assumes an important role of mitochondria in mediating the normal tissue and tumor response to FLASH dose rates.
Collapse
|
45
|
Kim LN, Rubenstein RN, Chu JJ, Allen RJ, Mehrara BJ, Nelson JA. Noninvasive Systemic Modalities for Prevention of Head and Neck Radiation-Associated Soft Tissue Injury: A Narrative Review. J Reconstr Microsurg 2022; 38:621-629. [PMID: 35213927 PMCID: PMC9402815 DOI: 10.1055/s-0042-1742731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Radiation-associated soft tissue injury is a potentially devastating complication for head and neck cancer patients. The damage can range from minor sequelae such as xerostomia, which requires frequent daily maintenance, to destructive degenerative processes such as osteoradionecrosis, which can contribute to flap failure and delay or reverse oral rehabilitation. Despite the need for effective radioprotectants, the literature remains sparse, primarily focused on interventions beyond the surgeon's control, such as maintenance of good oral hygiene or modulation of radiation dose. METHODS This narrative review aggregates and explores noninvasive, systemic treatment modalities for prevention or amelioration of radiation-associated soft tissue injury. RESULTS We highlighted nine modalities with the most clinical potential, which include amifostine, melatonin, palifermin, hyperbaric oxygen therapy, photobiomodulation, pentoxifylline-tocopherol-clodronate, pravastatin, transforming growth factor-β modulators, and deferoxamine, and reviewed the benefits and limitations of each modality. Unfortunately, none of these modalities are supported by strong evidence for prophylaxis against radiation-associated soft tissue injury. CONCLUSION While we cannot endorse any of these nine modalities for immediate clinical use, they may prove fruitful areas for further investigation.
Collapse
Affiliation(s)
- Leslie N. Kim
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robyn N. Rubenstein
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jacqueline J. Chu
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J. Allen
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Babak J. Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonas A. Nelson
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
46
|
Baic A, Plaza D, Lange B, Michalecki Ł, Stanek A, Kowalczyk A, Ślosarek K, Cholewka A. Long-Term Skin Temperature Changes after Breast Cancer Radiotherapy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:6891. [PMID: 35682472 PMCID: PMC9180487 DOI: 10.3390/ijerph19116891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/16/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022]
Abstract
The aim of the study was to use thermal imaging to evaluate long-term chest temperature changes in patients who had previously been treated with radiotherapy. The examination with a thermal imaging camera involved 144 women-48 of them were patients after RT, 48 were females before breast cancer radiotherapy and the last group of participants were 48 healthy women. All patients (before and after radiotherapy) were divided into women after mastectomy and those after conservative surgery. In addition, the first group of women, those who had received radiotherapy, were divided into three other groups: up to 1 year after RT, over 1 year and up to 5 years after RT and over 5 years after RT. Due to this, it was possible to compare the results and analyse the differences between the temperature in the healthy and treated breasts. The comparison of obtained temperature results showed that the area treated by ionizing radiation is characterized by a higher temperature even a few years after the finished treatment. It is worth mentioning that despite the fact that the difference was visible on the thermograms, the patients had no observable skin lesion or change in color at the treatment site. For the results of the study provided for the group of healthy patients, there were no significant differences observed between the average temperatures in the breasts. The use of thermal imaging in the evaluation of skin temperature changes after radiotherapy showed that the average temperature in the treated breast area can change even a long time after treatment.
Collapse
Affiliation(s)
- Agnieszka Baic
- Faculty of Science and Technology, University of Silesia, 75 Pułku Piechoty Street 1A, 41-500 Chorzów, Poland;
| | - Dominika Plaza
- Radiotherapy Planning Department, Maria Skłodowska—Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej Street 15, 44-102 Gliwice, Poland; (D.P.); (K.Ś.)
| | - Barbara Lange
- IIIrd Radiotherapy and Chemotherapy Department, Maria Skłodowska—Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej Street 15, 44-102 Gliwice, Poland;
| | - Łukasz Michalecki
- Department of Radiation Oncology, University Clinical Center, Medical University of Silesia, Ceglana Street 35, 40-514 Katowice, Poland;
| | - Agata Stanek
- Clinical Department of Internal Medicine, Angiology and Physical Medicine, Medical University of Silesia, Poniatowskiego Steet 15, 40-055 Katowice, Poland;
| | - Anna Kowalczyk
- Department of Physiotherapy, School of Health Sciences, Medical University of Silesia, Medyków Street 12, 40-752 Katowice, Poland;
| | - Krzysztof Ślosarek
- Radiotherapy Planning Department, Maria Skłodowska—Curie National Research Institute of Oncology Gliwice Branch, Wybrzeze Armii Krajowej Street 15, 44-102 Gliwice, Poland; (D.P.); (K.Ś.)
| | - Armand Cholewka
- Faculty of Science and Technology, University of Silesia, 75 Pułku Piechoty Street 1A, 41-500 Chorzów, Poland;
| |
Collapse
|
47
|
P144 a Transforming Growth Factor Beta Inhibitor Peptide, Generates Antifibrogenic Effects in a Radiotherapy Induced Fibrosis Model. Curr Oncol 2022; 29:2650-2661. [PMID: 35448191 PMCID: PMC9024500 DOI: 10.3390/curroncol29040217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/04/2022] Open
Abstract
Radiation-induced fibrosis (RIF) is a severe side effect related with soft tissues sarcomas (STS) radiotherapy. RIF is a multicellular process initiated primarily by TGF-β1 that is increased in irradiated tissue, whose signaling leads to intracellular Smad2/3 phosphorylation and further induction of profibrotic target genes. P144 (Disetertide©) is a peptide inhibitor of TGF-β1 and is proposed as a candidate compound for reducing RIF associated wound healing problems and muscle fibrosis in STS. Methods: A treatment and control group of WNZ rabbits were employed to implement a brachytherapy animal model, through catheter implantation at the lower limb. Two days after implantation, animals received 20 Gy isodosis, intended to induce a high RIF grade. The treatment group received intravenous P144 administration following a brachytherapy session, repeated at 24–72 h post-radiation, while the control group received placebo. Four weeks later, affected muscular tissues underwent histological processing for collagen quantification and P-Smad2/3 immunohistochemistry through image analysis. Results: High isodosis Brachytherapy produced remarkable fibrosis in this experimental model. Results showed retained macro and microscopical morphology of muscle in the P144 treated group, with reduced extracellular matrix fibrosis, with a lower area of collagen deposition measured through Masson’s trichrome staining. Intravenous P144 also induced a significant reduction in Smad2/3 phosphorylation levels compared with the placebo group. Conclusions: P144 administration clearly reduces RIF and opens a new potential co-treatment approach to reduce complications in soft tissue sarcoma (STS) radiotherapy. Further studies are required to establish whether the dosage and timing optimization of P144 administration, in different RIF phases, might entirely avoid fibrosis associated with STS brachytherapy.
Collapse
|
48
|
In Vitro and In Vivo Cardioprotective Effects of Curcumin against Doxorubicin-Induced Cardiotoxicity: A Systematic Review. JOURNAL OF ONCOLOGY 2022; 2022:7277562. [PMID: 35237323 PMCID: PMC8885194 DOI: 10.1155/2022/7277562] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022]
Abstract
Objective This study aimed to review the potential chemoprotective effects of curcumin against the doxorubicin-induced cardiotoxicity. Methods According to the PRISMA guideline, a comprehensive systematic search was performed in different electronic databases (Web of Science, PubMed, and Scopus) up to July 2021. One hundred and sixty-four studies were screened in accordance with a predefined set of inclusion and exclusion criteria. Eighteen eligible articles were finally included in the current systematic review. Results According to the in vitro and in vivo findings, it was found that doxorubicin administration leads to decreased cell survival, increased mortality, decreased bodyweight, heart weight, and heart to the bodyweight ratio compared to the control groups. However, curcumin cotreatment demonstrated an opposite pattern in comparison with the doxorubicin-treated groups alone. Other findings showed that doxorubicin significantly induces biochemical changes in the cardiac cells/tissue. Furthermore, the histological changes on the cardiac tissue were observed following doxorubicin treatment. Nevertheless, for most of the cases, these biochemical and histological changes mediated by doxorubicin were reversed near to control groups following curcumin coadministration. Conclusion It can be mentioned that coadministration of curcumin alleviates the doxorubicin-induced cardiotoxicity. Curcumin exerts these cardioprotective effects through different mechanisms of antioxidant, antiapoptosis, and anti-inflammatory. Since the finding presented in this systematic review are based on in vitro and in vivo studies, suggesting the use of curcumin in cancer patients as a cardioprotector agent against cardiotoxicity mediated by doxorubicin requires further clinical studies.
Collapse
|
49
|
Müller-Seubert W, Ostermaier P, Horch RE, Distel L, Frey B, Cai A, Arkudas A. Intra- and Early Postoperative Evaluation of Malperfused Areas in an Irradiated Random Pattern Skin Flap Model Using Indocyanine Green Angiography and Near-Infrared Reflectance-Based Imaging and Infrared Thermography. J Pers Med 2022; 12:jpm12020237. [PMID: 35207725 PMCID: PMC8880010 DOI: 10.3390/jpm12020237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Background: Assessment of tissue perfusion after irradiation of random pattern flaps still remains a challenge. Methods: Twenty-five rats received harvesting of bilateral random pattern fasciocutaneous flaps. Group 1 served as nonirradiated control group. The right flaps of the groups 2–5 were irradiated with 20 Gy postoperatively (group 2), 3 × 12 Gy postoperatively (group 3), 20 Gy preoperatively (group 4) and 3 × 12 Gy preoperatively (group 5). Imaging with infrared thermography, indocyanine green angiography and near-infrared reflectance-based imaging were performed to detect necrotic areas of the flaps. Results: Analysis of the percentage of the necrotic area of the irradiated flaps showed a statistically significant increase from day 1 to 14 only in group 5 (p < 0.05). Indocyanine green angiography showed no differences (p > 0.05) of the percentage of the nonperfused area between all days in group 1 and 3, but a decrease in group 2 in both the left and the right flaps. Infrared thermography and near-infrared reflectance-based imaging did not show evaluable differences. Conclusion: Indocyanine green angiography is more precise in prediction of necrotic areas in random pattern skin flaps when compared to hyperspectral imaging, thermography or clinical impression. Preoperative fractional irradiation with a lower individual dose but a higher total dose has a more negative impact on flap perfusion compared to higher single stage irradiation.
Collapse
Affiliation(s)
- Wibke Müller-Seubert
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany; (P.O.); (R.E.H.); (A.C.); (A.A.)
- Correspondence: ; Tel.: +49-9131-85-33296; Fax: +49-9131-85-39327
| | - Patrick Ostermaier
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany; (P.O.); (R.E.H.); (A.C.); (A.A.)
| | - Raymund E. Horch
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany; (P.O.); (R.E.H.); (A.C.); (A.A.)
| | - Luitpold Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany;
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany;
| | - Aijia Cai
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany; (P.O.); (R.E.H.); (A.C.); (A.A.)
| | - Andreas Arkudas
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nuremberg FAU, 91054 Erlangen, Germany; (P.O.); (R.E.H.); (A.C.); (A.A.)
| |
Collapse
|
50
|
Ramia P, Bodgi L, Mahmoud D, Mohammad MA, Youssef B, Kopek N, Al-Shamsi H, Dagher M, Abu-Gheida I. Radiation-Induced Fibrosis in Patients with Head and Neck Cancer: A Review of Pathogenesis and Clinical Outcomes. CLINICAL MEDICINE INSIGHTS: ONCOLOGY 2022; 16:11795549211036898. [PMID: 35125900 PMCID: PMC8808018 DOI: 10.1177/11795549211036898] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
Radiotherapy-related fibrosis remains one of the most challenging treatment related side effects encountered by patients with head and neck cancer. Several established and ongoing novel therapies have been studied with paucity of data in how to best treat these patients. This review aims to provide researchers and health care providers with a comprehensive review on the presentation, etiology, and therapeutic options for this serious condition.
Collapse
Affiliation(s)
- Paul Ramia
- McGill University Health Centre, Montreal, QC, Canada
| | - Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Dima Mahmoud
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammad A Mohammad
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Bassem Youssef
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Neil Kopek
- McGill University Health Centre, Montreal, QC, Canada
| | - Humaid Al-Shamsi
- Burjeel Cancer Institute, Abu-Dhabi, United Arab Emirates.,Emirates Oncology Society, Dubai, United Arab Emirates.,University of Sharjah, Sharjah, United Arab Emirates
| | - Mona Dagher
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ibrahim Abu-Gheida
- Burjeel Cancer Institute, Abu-Dhabi, United Arab Emirates.,Emirates Oncology Society, Dubai, United Arab Emirates.,United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|