1
|
Chu H, de Vette SPM, Neh H, Sijtsema NM, Steenbakkers RJHM, Moreno A, Langendijk JA, van Ooijen PMA, Fuller CD, van Dijk LV. Three-Dimensional Deep Learning Normal Tissue Complication Probability Model to Predict Late Xerostomia in Patients With Head and Neck Cancer. Int J Radiat Oncol Biol Phys 2025; 121:269-280. [PMID: 39147208 PMCID: PMC11646177 DOI: 10.1016/j.ijrobp.2024.07.2334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Conventional normal tissue complication probability (NTCP) models for patients with head and neck cancer are typically based on single-value variables, which, for radiation-induced xerostomia, are baseline xerostomia and mean salivary gland doses. This study aimed to improve the prediction of late xerostomia by using 3-dimensional information from radiation dose distributions, computed tomography imaging, organ-at-risk segmentations, and clinical variables with deep learning (DL). METHODS AND MATERIALS An international cohort of 1208 patients with head and neck cancer from 2 institutes was used to train and twice validate DL models (deep convolutional neural network, EfficientNet-v2, and ResNet) with 3-dimensional dose distribution, computed tomography scan, organ-at-risk segmentations, baseline xerostomia score, sex, and age as input. The NTCP endpoint was moderate-to-severe xerostomia 12 months postradiation therapy. The DL models' prediction performance was compared with a reference model: a recently published xerostomia NTCP model that used baseline xerostomia score and mean salivary gland doses as input. Attention maps were created to visualize the focus regions of the DL predictions. Transfer learning was conducted to improve the DL model performance on the external validation set. RESULTS All DL-based NTCP models showed better performance (area under the receiver operating characteristic curve [AUC]test, 0.78-0.79) than the reference NTCP model (AUCtest, 0.74) in the independent test. Attention maps showed that the DL model focused on the major salivary glands, particularly the stem cell-rich region of the parotid glands. DL models obtained lower external validation performance (AUCexternal, 0.63) than the reference model (AUCexternal, 0.66). After transfer learning on a small external subset, the DL model (AUCtl, external, 0.66) performed better than the reference model (AUCtl, external, 0.64). CONCLUSION DL-based NTCP models performed better than the reference model when validated in data from the same institute. Improved performance in the external data set was achieved with transfer learning, demonstrating the need for multicenter training data to realize generalizable DL-based NTCP models.
Collapse
Affiliation(s)
- Hung Chu
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Suzanne P M de Vette
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hendrike Neh
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nanna M Sijtsema
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Roel J H M Steenbakkers
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Amy Moreno
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Johannes A Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter M A van Ooijen
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Clifton D Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lisanne V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
2
|
Starska-Kowarska K. Role of Mesenchymal Stem/Stromal Cells in Head and Neck Cancer-Regulatory Mechanisms of Tumorigenic and Immune Activity, Chemotherapy Resistance, and Therapeutic Benefits of Stromal Cell-Based Pharmacological Strategies. Cells 2024; 13:1270. [PMID: 39120301 PMCID: PMC11311692 DOI: 10.3390/cells13151270] [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: 06/08/2024] [Revised: 07/11/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Head and neck cancer (HNC) entails a heterogenous neoplastic disease that arises from the mucosal epithelium of the upper respiratory system and the gastrointestinal tract. It is characterized by high morbidity and mortality, being the eighth most common cancer worldwide. It is believed that the mesenchymal/stem stromal cells (MSCs) present in the tumour milieu play a key role in the modulation of tumour initiation, development and patient outcomes; they also influence the resistance to cisplatin-based chemotherapy, the gold standard for advanced HNC. MSCs are multipotent, heterogeneous and mobile cells. Although no MSC-specific markers exist, they can be recognized based on several others, such as CD73, CD90 and CD105, while lacking the presence of CD45, CD34, CD14 or CD11b, CD79α, or CD19 and HLA-DR antigens; they share phenotypic similarity with stromal cells and their capacity to differentiate into other cell types. In the tumour niche, MSC populations are characterized by cell quiescence, self-renewal capacity, low reactive oxygen species production and the acquisition of epithelial-to-mesenchymal transition properties. They may play a key role in the process of acquiring drug resistance and thus in treatment failure. The present narrative review examines the links between MSCs and HNC, as well as the different mechanisms involved in the development of resistance to current chemo-radiotherapies in HNC. It also examines the possibilities of pharmacological targeting of stemness-related chemoresistance in HNSCC. It describes promising new strategies to optimize chemoradiotherapy, with the potential to personalize patient treatment approaches, and highlights future therapeutic perspectives in HNC.
Collapse
Affiliation(s)
- Katarzyna Starska-Kowarska
- Department of Physiology, Pathophysiology and Clinical Immunology, Department of Clinical Physiology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland; ; Tel.: +48-42-2725237
- Department of Otorhinolaryngology, EnelMed Center Expert, Lodz, Drewnowska 58, 91-001 Lodz, Poland
| |
Collapse
|
3
|
Carlander ALF, Gundestrup AK, Jansson PM, Follin B, Hoeeg C, Kousholt BS, Larsen RT, Jakobsen KK, Rimborg S, Fischer-Nielsen A, Grønhøj C, Buchwald CV, Lynggaard CD. Mesenchymal Stromal/Stem Cell Therapy Improves Salivary Flow Rate in Radiation-Induced Salivary Gland Hypofunction in Preclinical in vivo Models: A Systematic Review and Meta-Analysis. Stem Cell Rev Rep 2024; 20:1078-1092. [PMID: 38430363 PMCID: PMC11087340 DOI: 10.1007/s12015-024-10700-y] [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: 02/17/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) have been suggested for salivary gland (SG) restoration following radio-induced salivary gland damage. This study aimed to determine the safety and effectiveness of MSC therapy on radio-induced SG damage and hypofunction in preclinical in vivo studies. METHODS PubMed and EMBASE were systematically searched for preclinical in vivo interventional studies evaluating efficacy and safety of MSC treatment following radio-induced salivary gland damage published before 10th of January 2022. The primary endpoint was salivary flow rate (SFR) evaluated in a meta-analysis. The study protocol was published and registered on PROSPERO ( www.crd.ac.uk/prospero ), registration number CRD42021227336. RESULTS A total of 16 preclinical in vivo studies were included for qualitative analysis (858 experimental animals) and 13 in the meta-analysis (404 experimental animals). MSCs originated from bone marrow (four studies), adipose tissue (10 studies) and salivary gland tissue (two studies) and were administered intravenously (three studies), intra-glandularly (11 studies) or subcutaneously (one study). No serious adverse events were reported. The overall effect on SFR was significantly increased with a standardized mean difference (SMD) of 6.99 (95% CI: 2.55-11.42). Studies reported improvements in acinar tissue, vascular areas and paracrine factors. CONCLUSION In conclusion, this systematic review and meta-analysis showed a significant effect of MSC therapy for restoring SG functioning and regenerating SG tissue following radiotherapy in preclinical in vivo studies without serious adverse events. MSC therapy holds significant therapeutic potential in the treatment of radio-induced xerostomia, but comprehensive, randomized, clinical trials in humans are required to ascertain their efficacy in a clinical setting.
Collapse
Affiliation(s)
- Amanda-Louise Fenger Carlander
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
- Department of Otolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, Copenhagen University hospital, Copenhagen, Denmark.
| | - Anders Kierkegaard Gundestrup
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Per Marcus Jansson
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Bjarke Follin
- Cardiology Stem Cell Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Cecilie Hoeeg
- Cardiology Stem Cell Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Birgitte Saima Kousholt
- Department of Clinical Medicine, Aarhus University Group for Understanding Systematic Reviews and Meta analyses in Translational Preclinical Science, Aarhus University, Copenhagen, Denmark
| | - Rasmus Tolstrup Larsen
- Department of Occupational Therapy and Physiotherapy, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Section of Social Medicine, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Kathrine Kronberg Jakobsen
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Susie Rimborg
- The Royal Danish Library, Copenhagen University Library, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Department of Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Grønhøj
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian von Buchwald
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Duch Lynggaard
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
4
|
Tam VCW, Ching JCF, Yip SST, Kwong VHY, Chan CPL, Wong KCW, Lee SWY. Examining patient-reported late toxicity and its association with quality of life and unmet need for symptom management among nasopharyngeal cancer survivors: a cross-sectional survey. Front Oncol 2024; 14:1378973. [PMID: 38694788 PMCID: PMC11061844 DOI: 10.3389/fonc.2024.1378973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/26/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction Alongside the improved survival of nasopharyngeal cancer (NPC), late radiation toxicities are alarmingly hampering survivors' quality of life. A patient-reported symptom burden survey is lacking to address the unmet need for symptom management among local NPC survivors. Methods A single-center cross-sectional survey was conducted on 211 NPC survivors who had completed radiation therapy for three to 120 months. We employed the Chinese version M. D. Anderson Symptom Inventory - Head & Neck Module (MDASI-HN-C), Functional Assessment of Cancer Therapy - Head & Neck (FACT-HN-C), and a question extracted from the Cancer Survivors' Unmet Needs Measure (CaSUN). Results Two hundred valid responses were collected. Participants suffered from at least four moderate to severe symptoms (mean = 4.84, SD = 4.99). The top five severe symptoms were dry mouth, mucus problems, difficulty swallowing or chewing, teeth or gum problems, and memory problems. MDASI-HN-C subscales were negatively correlated with the physical, emotional, functional, and HN-specific domains of the FACT-HN-C. The unmet need for symptom management was positively associated with symptom burden, either general symptoms (Adjusted odds ratio [ORadj] = 1.566, 95% CI = 1.282 - 1.914, p < 0.001) or top-5 symptoms (ORadj = 1.379, 95% CI = 1.185 - 1.604, p < 0.001), while negatively associated with post-RT time (ORadj = 0.981, 95% CI [0.972, 0.991], p < 0.001). Conclusion Virtually all NPC survivors suffer from late toxicities, which interplay with survivors' perceptions intricately to affect their unmet needs for symptom management. Personalized supportive care strategies with regular assessments and stratifications are warranted.
Collapse
Affiliation(s)
- Victor C. W. Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, Hong Kong SAR, China
| | - Jerry C. F. Ching
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Sindy S. T. Yip
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Virginia H. Y. Kwong
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, Hong Kong SAR, China
| | - Catherine P. L. Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kenneth C. W. Wong
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, Hong Kong SAR, China
| | - Shara W. Y. Lee
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Soto-Gamez A, van Es M, Hageman E, Serna-Salas SA, Moshage H, Demaria M, Pringle S, Coppes RP. Mesenchymal stem cell-derived HGF attenuates radiation-induced senescence in salivary glands via compensatory proliferation. Radiother Oncol 2024; 190:109984. [PMID: 37926332 DOI: 10.1016/j.radonc.2023.109984] [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: 06/15/2023] [Revised: 10/21/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND & AIM Irradiation of the salivary glands during head and neck cancer treatment induces cellular senescence in response to DNA damage and contributes to radiation-induced hyposalivation by affecting the salivary gland stem/progenitor cell (SGSC) niche. Cellular senescence, such as that induced by radiation, is a state of cell-cycle arrest, accompanied by an altered pro-inflammatory secretome known as the senescence-associated secretory phenotype (SASP) with potential detrimental effects on the surrounding microenvironment. We hypothesized that the pro-regenerative properties of mesenchymal stem cells (MSCs) may attenuate cellular senescence post-irradiation. Therefore, here we evaluated the effects of adipose-derived MSCs (ADSCs) on the radiation-induced response of salivary gland organoids (SGOs). METHODS Proteomic analyses to identify soluble mediators released by ADSCs co-cultured with SGOS revealed secretion of hepatocyte growth factor (HGF) in ADSCs, suggesting a possible role in the stem cell crosstalk. Next, the effect of recombinant HGF in the culture media of ex vivo grown salivary gland cells was tested in 2D monolayers and 3D organoid models. RESULTS Treatment with HGF robustly increased salivary gland cell proliferation. Importantly, HGF supplementation post-irradiation enhanced proliferation at lower doses of radiation (0, 3, 7 Gy), but not at higher doses (10, 14 Gy) where most cells stained positive for senescence-associated beta-galactosidase. Furthermore, HGF had no effect on the senescence-associated secretory phenotype (SASP) of irradiated SGOs, suggesting there may be compensatory proliferation by cell-division competent cells instead of a reversal of cellular senescence after irradiation. CONCLUSION ADSCs may positively influence radiation recovery through HGF secretion and can promote the ex vivo expansion of salivary gland stem/progenitor cells to enhance the effects of co-transplanted SGSC.
Collapse
Affiliation(s)
- A Soto-Gamez
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - M van Es
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - E Hageman
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - S A Serna-Salas
- Dept of Gastroenterology and Hepatology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - H Moshage
- Dept of Gastroenterology and Hepatology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - M Demaria
- European Research Institute for the Biology of Ageing, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - S Pringle
- Dept. of Rheumatology and Clinical Immunology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - R P Coppes
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands.
| |
Collapse
|
6
|
Lyons KM, Cannon RD, Beumer J, Bakr MM, Love RM. Microbial Analysis of Obturators During Maxillofacial Prosthodontic Treatment Over an 8-Year Period. Cleft Palate Craniofac J 2023; 60:1426-1441. [PMID: 35642284 DOI: 10.1177/10556656221104940] [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: 11/17/2022] Open
Abstract
The aim of the study was to investigate the microbial colonization (by Candida species, anaerobic and facultative anaerobic bacteria) of maxillary obturators used for the restoration of maxillary defects, including during radiotherapy. Retrospective cohort study. Fifteen patients requiring a maxillary obturator prosthesis had swabs of their obturators and adjacent tissues taken at different stages of their treatment over a period of 8 years. Identification of microbial species from the swabs was carried out using randomly amplified polymorphic DNA polymerase chain reaction (RAPD PCR) analysis, checkerboard DNA-DNA hybridization, CHROMagar Candida chromogenic agar, and DNA sequencing. Candida species were detected in all patients and all patients developed mucositis and candidiasis during radiotherapy which was associated with an increase in colonization of surfaces with Candida spp., particularly C albicans. Microbial colonization increased during radiotherapy and as an obturator aged, and decreased following a reline, delivery of a new prosthesis, or antifungal treatment during radiotherapy. Microbial colonization of maxillary obturators was related to the stage of treatment, age of the obturator material, radiotherapy and antifungal medications, and antifungal treatment may be recommended if C albicans colonization of palatal tissues is greater than 105 colony-forming units per cm2 following the first week of radiotherapy.
Collapse
Affiliation(s)
- Karl M Lyons
- Department of Oral Rehabilitation and Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Richard D Cannon
- Department of Oral Sciences and Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - John Beumer
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Mahmoud M Bakr
- School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Robert M Love
- School of Medicine and Dentistry, Griffith University, Queensland, Australia
| |
Collapse
|
7
|
Marinkovic M, Tran ON, Wang H, Abdul-Azees P, Dean DD, Chen XD, Yeh CK. Autologous mesenchymal stem cells offer a new paradigm for salivary gland regeneration. Int J Oral Sci 2023; 15:18. [PMID: 37165024 PMCID: PMC10172302 DOI: 10.1038/s41368-023-00224-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/20/2023] [Accepted: 03/29/2023] [Indexed: 05/12/2023] Open
Abstract
Salivary gland (SG) dysfunction, due to radiotherapy, disease, or aging, is a clinical manifestation that has the potential to cause severe oral and/or systemic diseases and compromise quality of life. Currently, the standard-of-care for this condition remains palliative. A variety of approaches have been employed to restore saliva production, but they have largely failed due to damage to both secretory cells and the extracellular matrix (niche). Transplantation of allogeneic cells from healthy donors has been suggested as a potential solution, but no definitive population of SG stem cells, capable of regenerating the gland, has been identified. Alternatively, mesenchymal stem cells (MSCs) are abundant, well characterized, and during SG development/homeostasis engage in signaling crosstalk with the SG epithelium. Further, the trans-differentiation potential of these cells and their ability to regenerate SG tissues have been demonstrated. However, recent findings suggest that the "immuno-privileged" status of allogeneic adult MSCs may not reflect their status post-transplantation. In contrast, autologous MSCs can be recovered from healthy tissues and do not present a challenge to the recipient's immune system. With recent advances in our ability to expand MSCs in vitro on tissue-specific matrices, autologous MSCs may offer a new therapeutic paradigm for restoration of SG function.
Collapse
Affiliation(s)
- Milos Marinkovic
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Olivia N Tran
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hanzhou Wang
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Parveez Abdul-Azees
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - David D Dean
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Xiao-Dong Chen
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA.
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, USA.
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
8
|
Zhou L, Zheng W, Huang S, Yang X. Integrated radiomics, dose-volume histogram criteria and clinical features for early prediction of saliva amount reduction after radiotherapy in nasopharyngeal cancer patients. Discov Oncol 2022; 13:145. [PMID: 36581739 PMCID: PMC9800672 DOI: 10.1007/s12672-022-00606-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Previously, the evaluation of xerostomia depended on subjective grading systems, rather than the accurate saliva amount reduction. Our aim was to quantify acute xerostomia with reduced saliva amount, and apply radiomics, dose-volume histogram (DVH) criteria and clinical features to predict saliva amount reduction by machine learning techniques. MATERIAL AND METHODS Computed tomography (CT) of parotid glands, DVH, and clinical data of 52 patients were collected to extract radiomics, DVH criteria and clinical features, respectively. Firstly, radiomics, DVH criteria and clinical features were divided into 3 groups for feature selection, in order to alleviate the masking effect of the number of features in different groups. Secondly, the top features in the 3 groups composed integrated features, and features selection was performed again for integrated features. In this study, feature selection was used as a combination of eXtreme Gradient Boosting (XGBoost) and SHapley Additive exPlanations (SHAP) to alleviate multicollinearity. Finally, 6 machine learning techniques were used for predicting saliva amount reduction. Meanwhile, top radiomics features were modeled using the same machine learning techniques for comparison. RESULT 17 integrated features (10 radiomics, 4 clinical, 3 DVH criteria) were selected to predict saliva amount reduction, with a mean square error (MSE) of 0.6994 and a R2 score of 0.9815. Top 17 and 10 selected radiomics features predicted saliva amount reduction, with MSE of 0.7376, 0.7519, and R2 score of 0.9805, 0.9801, respectively. CONCLUSION With the same number of features, integrated features (radiomics + DVH criteria + clinical) performed better than radiomics features alone. The important DVH criteria and clinical features mainly included, white blood cells (WBC), parotid_glands_Dmax, Age, parotid_glands_V15, hemoglobin (Hb), BMI and parotid_glands_V45.
Collapse
Affiliation(s)
- Lang Zhou
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- Department of Biomedical Engineering, South China University of Technology, Guangzhou, 510640, Guangdong Province, China
| | - Wanjia Zheng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- Department of Radiation Oncology, Southern Theater Air Force Hospital of the People's Liberation Army, Guangzhou, 510050, Guangdong Province, China
| | - Sijuan Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| | - Xin Yang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| |
Collapse
|
9
|
Uchida H, Ingalls MH, Maruyama EO, Johnston CJ, Hernady E, Faustoferri RC, Ovitt CE. Short-term and bystander effects of radiation on murine submandibular glands. Dis Model Mech 2022; 15:dmm049570. [PMID: 36263624 PMCID: PMC9683099 DOI: 10.1242/dmm.049570] [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: 03/21/2022] [Accepted: 10/13/2022] [Indexed: 11/20/2022] Open
Abstract
Many patients treated for head and neck cancers experience salivary gland hypofunction due to radiation damage. Understanding the mechanisms of cellular damage induced by radiation treatment is important in order to design methods of radioprotection. In addition, it is crucial to recognize the indirect effects of irradiation and the systemic responses that may alter saliva secretion. In this study, radiation was delivered to murine submandibular glands (SMGs) bilaterally, using a 137Cs gamma ray irradiator, or unilaterally, using a small-animal radiation research platform (SARRP). Analysis at 3, 24 and 48 h showed dynamic changes in mRNA and protein expression in SMGs irradiated bilaterally. Unilateral irradiation using the SARRP caused similar changes in the irradiated SMGs, as well as significant off-target, bystander effects in the non-irradiated contralateral SMGs.
Collapse
Affiliation(s)
- Hitoshi Uchida
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Matthew H. Ingalls
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Eri O. Maruyama
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Carl J. Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Roberta C. Faustoferri
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Catherine E. Ovitt
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642USA
| |
Collapse
|
10
|
Robijns J, Nair RG, Lodewijckx J, Arany P, Barasch A, Bjordal JM, Bossi P, Chilles A, Corby PM, Epstein JB, Elad S, Fekrazad R, Fregnani ER, Genot MT, Ibarra AMC, Hamblin MR, Heiskanen V, Hu K, Klastersky J, Lalla R, Latifian S, Maiya A, Mebis J, Migliorati CA, Milstein DMJ, Murphy B, Raber-Durlacher JE, Roseboom HJ, Sonis S, Treister N, Zadik Y, Bensadoun RJ. Photobiomodulation therapy in management of cancer therapy-induced side effects: WALT position paper 2022. Front Oncol 2022; 12:927685. [PMID: 36110957 PMCID: PMC9468822 DOI: 10.3389/fonc.2022.927685] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
DisclaimerThis article is based on recommendations from the 12th WALT Congress, Nice, October 3-6, 2018, and a follow-up review of the existing data and the clinical observations of an international multidisciplinary panel of clinicians and researchers with expertise in the area of supportive care in cancer and/or PBM clinical application and dosimetry. This article is informational in nature. As with all clinical materials, this paper should be used with a clear understanding that continued research and practice could result in new insights and recommendations. The review reflects the collective opinion and, as such, does not necessarily represent the opinion of any individual author. In no event shall the authors be liable for any decision made or action taken in reliance on the proposed protocols.ObjectiveThis position paper reviews the potential prophylactic and therapeutic effects of photobiomodulation (PBM) on side effects of cancer therapy, including chemotherapy (CT), radiation therapy (RT), and hematopoietic stem cell transplantation (HSCT).BackgroundThere is a considerable body of evidence supporting the efficacy of PBM for preventing oral mucositis (OM) in patients undergoing RT for head and neck cancer (HNC), CT, or HSCT. This could enhance patients’ quality of life, adherence to the prescribed cancer therapy, and treatment outcomes while reducing the cost of cancer care.MethodsA literature review on PBM effectiveness and dosimetry considerations for managing certain complications of cancer therapy were conducted. A systematic review was conducted when numerous randomized controlled trials were available. Results were presented and discussed at an international consensus meeting at the World Association of photobiomoduLation Therapy (WALT) meeting in 2018 that included world expert oncologists, radiation oncologists, oral oncologists, and oral medicine professionals, physicists, engineers, and oncology researchers. The potential mechanism of action of PBM and evidence of PBM efficacy through reported outcomes for individual indications were assessed.ResultsThere is a large body of evidence demonstrating the efficacy of PBM for preventing OM in certain cancer patient populations, as recently outlined by the Multinational Association for Supportive Care in Cancer/International Society of Oral Oncology (MASCC/ISOO). Building on these, the WALT group outlines evidence and prescribed PBM treatment parameters for prophylactic and therapeutic use in supportive care for radiodermatitis, dysphagia, xerostomia, dysgeusia, trismus, mucosal and bone necrosis, lymphedema, hand-foot syndrome, alopecia, oral and dermatologic chronic graft-versus-host disease, voice/speech alterations, peripheral neuropathy, and late fibrosis amongst cancer survivors.ConclusionsThere is robust evidence for using PBM to prevent and treat a broad range of complications in cancer care. Specific clinical practice guidelines or evidence-based expert consensus recommendations are provided. These recommendations are aimed at improving the clinical utilization of PBM therapy in supportive cancer care and promoting research in this field. It is anticipated these guidelines will be revised periodically.
Collapse
Affiliation(s)
- Jolien Robijns
- UHasselt, Faculty of Medicine and Life Sciences, Diepenbeek, Belgium
| | - Raj G. Nair
- Oral Medicine, Oral Pathology and Oral Oncology, Griffith University, Department of Haematology and Oncology, Gold Coast University Hospital, Gold Coast, QL, Australia
| | - Joy Lodewijckx
- UHasselt, Faculty of Medicine and Life Sciences, Diepenbeek, Belgium
| | - Praveen Arany
- School of Dental Medicine, Oral Biology and Biomedical Engineering, University at Buffalo, Buffalo, NY, United States
| | - Andrei Barasch
- Harvard School of Dental Medicine, Division of Oral Medicine and Dentistry, Boston, MA, United States
| | - Jan M. Bjordal
- Physiotherapy Research Group, IGS, University of Bergen, Bergen, Norway
| | - Paolo Bossi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Anne Chilles
- Radiotherapy Department, Institut Curie, Paris, France
| | - Patricia M. Corby
- New York University College of Dentistry, Bluestone Center for Clinical Research, New York, NY, United States
| | - Joel B. Epstein
- City of Hope Duarte, CA and Cedars-Sinai Health System, Los Angeles, CA, United States
| | - Sharon Elad
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, United States
| | - Reza Fekrazad
- Department of Periodontology, Dental Faculty – Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
| | | | - Marie-Thérèse Genot
- Laser Therapy Unit, Institut Jules Bordet, Centre des Tumeurs de l’Université Libre de Bruxelles, Brussels, Belgium
| | - Ana M. C. Ibarra
- Postgraduate Program on Biophotonics Applied to Health Sciences, Nove de Julho University, São Paulo, Brazil
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Vladimir Heiskanen
- Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Ken Hu
- Department of Radiation Oncology, NYU Langone Health, New York, NY, United States
| | | | - Rajesh Lalla
- Section of Oral Medicine, University of Connecticut School of Dental Medicine, Farmington, CT, United States
| | - Sofia Latifian
- Department of Medicine, Institut Jules Bordet, Universiteí Libre de Bruxelles, Brussels, Belgium
| | - Arun Maiya
- Manipal College of Health Professions, MAHE, Manipal, India
| | - Jeroen Mebis
- UHasselt, Faculty of Medicine and Life Sciences, Diepenbeek, Belgium
| | - Cesar A. Migliorati
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, Florida, United States
| | - Dan M. J. Milstein
- Oral and Maxillofacial Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Barbara Murphy
- Department of Oncology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Judith E. Raber-Durlacher
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, Department of Oral Medicine, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, Netherlands
| | - Hendrik J. Roseboom
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, Department of Oral Medicine, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, Netherlands
| | - Stephen Sonis
- Division of Oral Medicine and Dentistry, Brigham and Women’s Hospital; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine; Division of Oral Medicine and Dentistry, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Nathaniel Treister
- Division of Oral Medicine and Dentistry, Brigham and Women’s Hospital; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine; Division of Oral Medicine and Dentistry, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Yehuda Zadik
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel, and Department of Oral Medicine, Sedation and Maxillofacial Imaging, Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - René-Jean Bensadoun
- Department of Radiation Oncology, Centre de Haute Energie, Nice, France
- *Correspondence: René-Jean Bensadoun,
| |
Collapse
|
11
|
Ahmed SF, Bakr NM, Abdelgawad N, Bashir DW. Possible radioprotection of submandibular glands in gamma-irradiated rats using kaempferol: a histopathological and immunohistochemical study. Int J Radiat Biol 2022; 99:396-405. [PMID: 35758986 DOI: 10.1080/09553002.2022.2094015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
BACKGROUND AND PURPOSE Salivary gland damage remains a problem despite advances in radiotherapy schedules for head and neck cancer. Kaempferol, a natural flavonoid, found in several fruits and vegetables, is a good antioxidant. This study was designed to evaluate the possible protective effects of kaempferol on submandibular glands (SMGs) of rats exposed to fractionated gamma irradiation. MATERIALS AND METHODS Twenty-four male adult Wistar albino rats were included in this study and assigned to three groups (n = 8). Rats in group K received kaempferol orally in five doses at a dose of 10 mg/kg/2 days for 10 days. Meanwhile, rats in group R were subjected to fractionated whole-body gamma irradiation at a dose of 2 Gy/5 days/week for 2 weeks (20 Gy), and the KR group received kaempferol as group K and then was subjected to a fractionated whole-body gamma irradiation as group R. SMG samples were collected on days 1 and 7 after the last radiation session; and processed for histopathological and immunohistochemical investigations. RESULTS The SMGs of group R showed focal atrophy and degeneration. Acini showed vacuolization and had pyknotic hyperchromatic nuclei. Striated ducts degenerated, shrunken, and were surrounded by empty spaces. The percentage of areas covered by cyclooxygenase-2 (COX-2) significantly increased, whereas the percentage of areas covered by proliferating cell nuclear antigen (PCNA) significantly decreased compared with those in group K. Cotreatment with kaempferol (group KR) partially preserved normal gland architecture where acinar vacuolation and degeneration were almost absent; however, some ducts degenerated. A significant decrease in the percentage of areas covered by COX-2 and a significant increase in the percentage of areas covered by PCNA were observed compared with those in group R. CONCLUSIONS Kaempferol has a possible radioprotective effect on the SMGs of rats exposed to fractionated gamma irradiation.
Collapse
Affiliation(s)
- Salwa Farid Ahmed
- Health Radiation Research Department, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Noura Mohammed Bakr
- Oral and Dental Biology Department, Faculty of Dental Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Nora Abdelgawad
- Oral Medicine, Periodontology, Diagnosis and Radiology Department, Faculty of Dental Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Dina W Bashir
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
Chao M, El Naqa I, Bakst RL, Lo YC, Peñagarícano JA. Cluster model incorporating heterogeneous dose distribution of partial parotid irradiation for radiotherapy induced xerostomia prediction with machine learning methods. Acta Oncol 2022; 61:842-848. [PMID: 35527717 DOI: 10.1080/0284186x.2022.2073187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
PURPOSE A cluster model incorporating heterogeneous dose distribution within the parotid gland was developed and validated retrospectively for radiotherapy (RT) induced xerostomia prediction with machine learning (ML) techniques. METHODS Sixty clusters were obtained at 1 Gy step size with threshold doses ranging from 1 to 60 Gy, for each of the enrolled 155 patients with HNC from three institutions. Feature clusters were selected with the neighborhood component analysis (NCA) and subsequently fed into four supervised ML models for xerostomia prediction comparison: support vector machines (SVM), k-nearest neighbor (kNN), naïve Bayes (NB), and random forest (RF). The predictive performance of each model was evaluated using cross validation resampling with the area-under-the-curves (AUC) of the receiver-operating-characteristic (ROC). The xerostomia predicting capacity using testing data was assessed with accuracy, sensitivity, and specificity for these models and three cluster connectivity choices. Mean dose based logistic regression served as the benchmark for evaluation. RESULTS Feature clusters identified by NCA fell in three threshold dose ranges: 5-15Gy, 25-35Gy, and 45-50Gy. Mean dose predictive power was 15% lower than that of the cluster model using the logistic regression classifier. Model validation demonstrated that kNN model outperformed slightly other three models but no substantial difference was observed. Applying the fine-tuned models to testing data yielded that the mean accuracy from SVM, kNN and NB models were between 0.68 and 0.7 while that of RF was ∼0.6. SVM model yielded the best sensitivity (0.76) and kNN model delivered consistent sensitivity and specificity. This is consistent with cross validation. Clusters calculated with three connectivity choices exhibited minimally different predictions. CONCLUSION Compared to mean dose, the proposed cluster model has shown its improvement as the xerostomia predictor. When combining with ML techniques, it could provide a clinically useful tool for xerostomia prediction and facilitate decision making during radiotherapy planning for patients with HNC.
Collapse
Affiliation(s)
- Ming Chao
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA
| | - Issam El Naqa
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Richard L. Bakst
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA
| | - Yeh-Chi Lo
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA
| | - José A. Peñagarícano
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
13
|
Jansson PM, Lynggaard CD, Carlander AF, Jensen SB, Follin B, Hoeeg C, Kousholt BS, Larsen RT, Grønhøj C, Jakobsen KK, Rimborg S, Fischer-Nielsen A, Menon JML, von Buchwald C. Mesenchymal stromal/stem cell therapy for radiation-induced salivary gland hypofunction in animal models: a protocol for a systematic review and meta-analysis. Syst Rev 2022; 11:72. [PMID: 35436971 PMCID: PMC9016929 DOI: 10.1186/s13643-022-01943-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/01/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Salivary gland (SG) hypofunction (objectively reduced saliva flow rate) and xerostomia (subjective sensation of dry mouth) are common and burdensome side effects of radiotherapy to the head and neck region. Currently, only sparse symptomatic treatment is available to ease the discomfort of xerostomia. The objective of this study is to assess the effect of mesenchymal stem cell (MSC) therapy on SG function after radiation-induced injury. METHODS This systematic review will include animal intervention studies assessing efficacy and safety of MSCs in treating radiation-induced SG hypofunction. The primary outcome is the effect of MSC administration on salivary flow rates (SFR), by comparing treated groups to control groups when available. Secondary outcomes are morphological and immunohistochemical effects as well as safety of MSC treatment. Electronic searches in MEDLINE (PubMed) and Embase databases will be constructed and validated according to the peer review of electronic search strategies (PRESS) and assessed by two independent researchers. Data from eligible studies will be extracted, pooled, and analyzed using random-effects models. Risk of bias will be evaluated with the Systematic Review Centre for Laboratory animal Experimentation (SYRCLE) risk of bias tool. DISCUSSION Thus far, critical appraisal of MSC therapy as an effective treatment for SG hypofunction caused solely by radiation injury has not been conducted. A summary of the existing literature on preclinical studies concerning this issue can provide valuable information about effectiveness, mode of action, and safety, allowing further optimization of preclinical and clinical trials. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42021227336.
Collapse
Affiliation(s)
- Per Marcus Jansson
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,The Royal Library, Library of Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Duch Lynggaard
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. .,The Royal Library, Library of Copenhagen, University of Copenhagen, Copenhagen, Denmark. .,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Amanda Fenger Carlander
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Siri Beier Jensen
- Deparment of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark
| | - Bjarke Follin
- Cardiology Stem Cell Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Cecilie Hoeeg
- Cardiology Stem Cell Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Birgitte Saima Kousholt
- Aarhus University Group for Understanding Systematic Reviews and Meta-analyses in Translational Preclinical Science, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rasmus Tolstrup Larsen
- Department of Occupational Therapy and Physiotherapy, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Section of Social Medicine, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Christian Grønhøj
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kathrine Kronberg Jakobsen
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Susie Rimborg
- The Royal Library, Library of Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Department of Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Julia M L Menon
- Systematic Review Center for Laboratory animal Experimentation (SYRCLE), Department for Health Evidence, Radboud University Medical Center, Nimegen, the Netherlands
| | - Christian von Buchwald
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
The Quantification of Salivary Flow and pH and Stomatognathic System Rehabilitation Interference in Patients with Oral Diseases, Post-Radiotherapy. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12083708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Xerostomia is a common complication post-radiotherapy in patients with oral cancer. The acute and long-term side effects can considerably reduce the patient’s quality of life. The aim of our study was to perform analysis of salivary flow and pH in patients after radiotherapy. Methodology: Clinical and laboratory evaluations were conducted in the 2014–2019 period; out of a total 58 subjects aged between 45 and 84, 28 individuals with oral cancer were selected from St. Spiridon Hospital, Clinic of Maxillo-facial Surgery and Oncology Hospital, Iasi post-radiotherapy. Results: Significant downsized mean values of the hydrogen ion concentration (pH) in saliva (p < 0.001) were recorded in patients after radiotherapy, pH value = 4.580 (±1.051). The mean value of resting salivary flow (MRSF) was significantly lower for the group of patients with radiotherapy (MRSF) = 0.145 mL/min. In 89.29% of cases (25 post-radiotherapy cases), in order to perform oral complex rehabilitation treatment, several endodontic and periodontal treatments were performed. A total of 78.57% of the cases received complex oral rehabilitation as mobile or hybrid prostheses or fixed solutions. Conclusion: Understanding post-radiotherapy salivary biochemic modifications in patients with oral cancer could be of critical importance, in view of related oral disorder prevention.
Collapse
|
15
|
Feng X, Wu Z, Xu J, Xu Y, Zhao B, Pang B, Qu X, Hu L, Hu L, Fan Z, Jin L, Xia D, Chang S, Wang J, Zhang C, Wang S. Dietary nitrate supplementation prevents radiotherapy-induced xerostomia. eLife 2021; 10:70710. [PMID: 34581269 PMCID: PMC8563005 DOI: 10.7554/elife.70710] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022] Open
Abstract
Management of salivary gland hypofunction caused by irradiation (IR) therapy for head and neck cancer remains lack of effective treatments. Salivary glands, especially the parotid gland, actively uptake dietary nitrate and secrete it into saliva. Here, we investigated the effect of dietary nitrate on the prevention and treatment of IR-induced parotid gland hypofunction in miniature pigs, and elucidated the underlying mechanism in human parotid gland cells. We found that nitrate administration prevented IR-induced parotid gland damage in a dose-dependent manner, by maintaining the function of irradiated parotid gland tissue. Nitrate could increase sialin expression, a nitrate transporter expressed in the parotid gland, making the nitrate-sialin feedback loop that facilitates nitrate influx into cells for maintaining cell proliferation and inhibiting apoptosis. Furthermore, nitrate enhanced cell proliferation via the epidermal growth factor receptor (EGFR)-protein kinase B (AKT)-mitogen-activated protein kinase (MAPK) signaling pathway in irradiated parotid gland tissue. Collectively, nitrate effectively prevented IR-induced xerostomia via the EGFR-AKT-MAPK signaling pathway. Dietary nitrate supplementation may provide a novel, safe, and effective way to resolve IR-induced xerostomia.
Collapse
Affiliation(s)
- Xiaoyu Feng
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.,Department of Pediatric Dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Zhifang Wu
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Junji Xu
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Yipu Xu
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Bin Zhao
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Baoxing Pang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Xingmin Qu
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Liang Hu
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Lei Hu
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhipeng Fan
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Luyuan Jin
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Dengsheng Xia
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Shimin Chang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Jingsong Wang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing, China
| | - Chunmei Zhang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Songlin Wang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.,Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Souza ACF, Monico-Neto M, Le Sueur-Maluf L, Pidone FAM, Antunes HKM, Ribeiro DA. Inflammatory activity and apoptosis are associated with tissue degeneration in the submandibular gland of rats submitted to paradoxical sleep deprivation. Odontology 2021; 110:278-286. [PMID: 34536167 DOI: 10.1007/s10266-021-00657-6] [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: 05/24/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
The aim of this study was to evaluate if paradoxical sleep deprivation is able to induce tissue degeneration, inflammatory activity and apoptosis in the submandibular gland of rats. A total of 24 male Wistar rats were distributed into the following groups: group 1-control (CTRL; n = 8): the animals were not submitted to any procedures; group 2-sleep deprivation (PS; n = 8): the animals were submitted to paradoxical sleep deprivation for 96 h and group 3-recovery (R; n = 8): the animals were submitted to sleep deprivation for 96 h, followed by a period of 96 h without any intervention. The following parameters were evaluated: histopathological analysis, immunohistochemistry for Ki-67, COX-2 and cleaved caspase-3 and gene expression of TNF-α, Interleukin 6 (IL-6), Interleukin 10 (IL-10) and cytochrome C by real-time PCR. The results pointed out cytoplasmic vacuoles and congested vessels in the parenchyma of submandibular gland the in PS and R groups. The expression of interleukins 6, 10 and TNF-ɑ was differentially expressed in the PS and R groups. Apoptosis was also triggered by means of increasing cleaved caspase-3 and cytochrome c expression. The cellular proliferation (Ki-67 index) was also positive in the R group. Taken together, our results demonstrate that sleep deprivation is capable of promoting tissue degeneration in the submandibular gland, as a result of inflammatory response and cellular death in rats.
Collapse
Affiliation(s)
- Ana Carolina Flygare Souza
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Marcos Monico-Neto
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Luciana Le Sueur-Maluf
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Flavia Andressa Mazzuco Pidone
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Hanna Karen Moreira Antunes
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil
| | - Daniel Araki Ribeiro
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, UNIFESP, Rua Silva Jardim, 136, Room 332, Vila Mathias, Santos, SP, 11050-020, Brazil.
| |
Collapse
|
17
|
Lim SB, Lee N, Zakeri K, Greer P, Fuangrod T, Coffman F, Cerviño L, Lovelock DM. Can the Risk of Dysphagia in Head and Neck Radiation Therapy Be Predicted by an Automated Transit Fluence Monitoring Process During Treatment? A First Comparative Study of Patient Reported Quality of Life and the Fluence-Based Decision Support Metric. Technol Cancer Res Treat 2021; 20:15330338211027906. [PMID: 34190006 PMCID: PMC8252347 DOI: 10.1177/15330338211027906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
PURPOSE/OBJECTIVE(S) The additional personnel and imaging procedures required for Adaptive Radiation Therapy (ART) pose a challenge for a broad implementation. We hypothesize that a change in transit fluence during the treatment course is correlated with the change of quality of life and thus can be used as a replanning trigger. MATERIALS/METHODS Twenty-one head and neck cancer (HNC) patients filled out an MD Anderson Dysphagia Inventory (MDADI) questionnaire, before-and-after the radiotherapy treatment course. The transit fluence was measured by the Watchdog (WD) in-vivo portal dosimetry system. The patients were monitored with daily WD and weekly CBCTs. The region of interest (ROI) of each patient was defined as the outer contour of the patient between approximate spine levels C1 to C4, essentially the neck and mandible inside the beam's eye view. The nth day integrated transit fluence change, Δϕn, and the volume change, ΔVROI, of the ROI of each patient was calculated from the corresponding WD and CBCT measurements. The correlation between MDADI scores and age, gender, planning mean dose to salivary glands <Dsg>, weight change ΔW, ΔVROI, and Δϕn, were analyzed using the ranked-Pearson correlation. RESULTS No statistically significant correlation was found for age, gender and ΔW. <Dsg> was found to have clinically important correlation with functional MDADI (ρ = -0.39, P = 0.081). ΔVROI was found to have statistically significant correlation of 0.44, 0.47 and 0.44 with global, physical and functional MDADI (P-value < 0.05). Δϕn was found to have statistically significant ranked-correlation (-0.46, -0.46 and -0.45) with physical, functional and total MDADI (P-value < 0.05). CONCLUSION A transit fluence based decision support metric (DSM) is statistically correlated with the dysphagia risk. It can not only be used as an early signal in assisting clinicians in the ART patient selection for replanning, but also lowers the resource barrier of ART implementation.
Collapse
Affiliation(s)
- Seng Boh Lim
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nancy Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kaveh Zakeri
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter Greer
- Calvary Mater Newcastle Hospital, New South Wales, Australia.,University of Newcastle, New South Wales, Australia
| | - Todsaporn Fuangrod
- HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | - Laura Cerviño
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - D Michael Lovelock
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
18
|
Pu D, Lee VHF, Chan KMK, Yuen MTY, Quon H, Tsang RKY. The Relationships Between Radiation Dosage and Long-term Swallowing Kinematics and Timing in Nasopharyngeal Carcinoma Survivors. Dysphagia 2021; 37:612-621. [PMID: 33909131 PMCID: PMC9072442 DOI: 10.1007/s00455-021-10311-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 04/20/2021] [Indexed: 11/16/2022]
Abstract
This study aimed to investigate the relationship between intensity-modulated radiation therapy (IMRT) dosimetry and swallowing kinematic and timing measures. Thirteen kinematic and timing measures of swallowing from videofluoroscopic analysis were used as outcome measures to reflect swallowing function. IMRT dosimetry was accessed for thirteen swallowing-related structures. A cohort of 44 nasopharyngeal carcinoma (NPC) survivors at least 3 years post-IMRT were recruited. The cohort had a mean age of 53.2 ± 11.9 years, 77.3% of whom were male. There was an average of 68.24 ± 14.15 months since end of IMRT; 41 (93.2%) had undergone concurrent chemotherapy. For displacement measures, female sex and higher doses to the cricopharyngeus, glottic larynx, and base of tongue were associated with reduced hyolaryngeal excursion and pharyngeal constriction, and more residue. For timing measures, higher dose to the genioglossus was associated with reduced processing time at all stages of the swallow. The inferior pharyngeal constrictor emerged with a distinctly different pattern of association with mean radiation dosage compared to other structures. Greater changes to swallowing kinematics and timing were observed for pudding thick consistency than thin liquid. Increasing radiation dosage to swallowing-related structures is associated with reduced swallowing kinematics. However, not all structures are affected the same way, therefore organ sparing during treatment planning for IMRT needs to consider function rather than focusing on select muscles. Dose-response relationships should be investigated with a comprehensive set of swallowing structures to capture the holistic process of swallowing.
Collapse
Affiliation(s)
- Dai Pu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Swallowing Research Laboratory, Faculty of Education, The University of Hong Kong, Hong Kong, China.,School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Sciences, Monash University, Frankston, Australia
| | - Victor H F Lee
- Department of Clinical Oncology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Karen M K Chan
- Swallowing Research Laboratory, Faculty of Education, The University of Hong Kong, Hong Kong, China
| | - Margaret T Y Yuen
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Oncology and Otolaryngology and Head and Neck Surgery, Johns Hopkins University, Baltimore, USA
| | - Raymond K Y Tsang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China. .,Department of Surgery and Department of ENT, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, China.
| |
Collapse
|
19
|
Tanaka J, Takamatsu K, Yukimori A, Kujiraoka S, Ishida S, Takakura I, Yasuhara R, Mishima K. Sox9 function in salivary gland development. J Oral Biosci 2021; 63:8-13. [PMID: 33497841 DOI: 10.1016/j.job.2021.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Organogenesis is regulated by morphogen signaling and transcription networks. These networks differ between organs, and identifying the organ-specific network is important to clarify the molecular mechanisms of development and regeneration of organs. Several studies have been conducted to identify salivary gland-specific networks using a mouse submandibular gland model. The submandibular glands (SMGs) of mice manifest as a thickening of the oral epithelium at embryonic day 11.5 and invaginate into the underlying mesenchyme. The network between Fgf10 and Sox9 is involved in SMG development in mice. HIGHLIGHT Sox9, a member of the Sox family, is expressed in the SMG in mice from the embryonic stage to the adult stage, although the distribution changes during development. A null mutation of mouse Sox9 is lethal during the neonatal period due to respiratory failure, whereas deletion of Sox9 in the oral epithelium using the Cre/lox P system, can lead to smaller initial buds of SMGs in conditional knockout (cKO) mice than in normal mice. In addition, we showed that adenoviral transduction of Sox9 and Foxc1 genes into mouse embryonic stem cell-derived oral ectoderm could induce salivary gland rudiment in an organoid culture system. ChIP-sequencing revealed that Sox9 possibly regulates several tube- and branching-formation-related genes. CONCLUSION Sox9 may serve as an essential transcription factor for salivary gland development. The Sox9-mediated pathway can be a promising candidate for regenerating damaged salivary glands.
Collapse
Affiliation(s)
- Junichi Tanaka
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Koki Takamatsu
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Akane Yukimori
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Satoko Kujiraoka
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Shoko Ishida
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Ikuko Takakura
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Rika Yasuhara
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| |
Collapse
|
20
|
Kushta AА, Shuvalov SМ. Techniques Of Restoring Swallowing Mechanisms In The Treatment Of Patients With Head And Neck Cancer: Postoperative Pain Relief, Plastic Surgery And Diet. RUSSIAN OPEN MEDICAL JOURNAL 2020. [DOI: 10.15275/rusomj.2020.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective — to review available publications and identify unresolved issues in assessing the functional state of the oral cavity and pharynx, along with swallowing disorders in oncopathology of head and neck, depending on the treatment method. The paper discusses the problems of the extent of surgical interventions and their relationship with the possibility of maintaining the act of swallowing, and contemporary understanding of the swallowing mechanism in normal and pathological conditions. Studies on postoperative pain management and feeding techniques of cancer patients are described. Conclusion — The mechanisms of impaired swallowing after surgery and chemoradiotherapy have been analyzed. For the first time, the mechanisms of damage to swallowing have been analyzed, and the ways of overcoming pathological conditions, such as dysphagia and pain, were substantiated, with topographic and anatomical details. The perspectives of resolving the issue of nutritional status restoration were outlined.
Collapse
|
21
|
Barrows CM, Wu D, Farach-Carson MC, Young S. Building a Functional Salivary Gland for Cell-Based Therapy: More than Secretory Epithelial Acini. Tissue Eng Part A 2020; 26:1332-1348. [PMID: 32829674 PMCID: PMC7759264 DOI: 10.1089/ten.tea.2020.0184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/20/2020] [Indexed: 11/13/2022] Open
Abstract
A few treatment options exist for patients experiencing xerostomia due to hyposalivation that occurs as a result of disease or injury to the gland. An opportunity for a permanent solution lies in the field of salivary gland replacement through tissue engineering. Recent success emboldens in the vision of producing a tissue-engineered salivary gland composed of differentiated salivary epithelial cells that are able to differentiate to form functional units that produce and deliver saliva to the oral cavity. This vision is augmented by advances in understanding cellular mechanisms that guide branching morphogenesis and salivary epithelial cell polarization in both acinar and ductal structures. Growth factors and other guidance cues introduced into engineered constructs help to develop a more complex glandular structure that seeks to mimic native salivary gland tissue. This review describes the separate epithelial phenotypes that make up the gland, and it describes their relationship with the other cell types such as nerve and vasculature that surround them. The review is organized around the links between the native components that form and contribute to various aspects of salivary gland development, structure, and function and how this information can drive the design of functional tissue-engineered constructs. In addition, we discuss the attributes of various biomaterials commonly used to drive function and form in engineered constructs. The review also contains a current description of the state-of-the-art of the field, including successes and challenges in creating materials for preclinical testing in animal models. The ability to integrate biomolecular cues in combination with a range of materials opens the door to the design of increasingly complex salivary gland structures that, once accomplished, can lead to breakthroughs in other fields of tissue engineering of epithelial-based exocrine glands or oral tissues.
Collapse
Affiliation(s)
- Caitlynn M.L. Barrows
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
- Department of Biosciences and Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Simon Young
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| |
Collapse
|
22
|
Saitou M, Gaylord EA, Xu E, May AJ, Neznanova L, Nathan S, Grawe A, Chang J, Ryan W, Ruhl S, Knox SM, Gokcumen O. Functional Specialization of Human Salivary Glands and Origins of Proteins Intrinsic to Human Saliva. Cell Rep 2020; 33:108402. [PMID: 33207190 PMCID: PMC7703872 DOI: 10.1016/j.celrep.2020.108402] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/31/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Salivary proteins are essential for maintaining health in the oral cavity and proximal digestive tract, and they serve as potential diagnostic markers for monitoring human health and disease. However, their precise organ origins remain unclear. Through transcriptomic analysis of major adult and fetal salivary glands and integration with the saliva proteome, the blood plasma proteome, and transcriptomes of 28+ organs, we link human saliva proteins to their source, identify salivary-gland-specific genes, and uncover fetal- and adult-specific gene repertoires. Our results also provide insights into the degree of gene retention during gland maturation and suggest that functional diversity among adult gland types is driven by specific dosage combinations of hundreds of transcriptional regulators rather than by a few gland-specific factors. Finally, we demonstrate the heterogeneity of the human acinar cell lineage. Our results pave the way for future investigations into glandular biology and pathology, as well as saliva's use as a diagnostic fluid.
Collapse
Affiliation(s)
- Marie Saitou
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, U.S.A; Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, U.S.A; Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Viken, Norway
| | - Eliza A Gaylord
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, CA, U.S.A
| | - Erica Xu
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, U.S.A
| | - Alison J May
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, CA, U.S.A
| | - Lubov Neznanova
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, NY, U.S.A
| | - Sara Nathan
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, CA, U.S.A
| | - Anissa Grawe
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, CA, U.S.A
| | - Jolie Chang
- Department of Otolaryngology, School of Medicine, University of California, San Francisco, CA, U.S.A
| | - William Ryan
- Department of Otolaryngology, School of Medicine, University of California, San Francisco, CA, U.S.A
| | - Stefan Ruhl
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, NY, U.S.A.
| | - Sarah M Knox
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, CA, U.S.A.
| | - Omer Gokcumen
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, U.S.A.
| |
Collapse
|
23
|
Luitje ME, Israel AK, Cummings MA, Giampoli EJ, Allen PD, Newlands SD, Ovitt CE. Long-Term Maintenance of Acinar Cells in Human Submandibular Glands After Radiation Therapy. Int J Radiat Oncol Biol Phys 2020; 109:1028-1039. [PMID: 33181249 DOI: 10.1016/j.ijrobp.2020.10.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE In a combined retrospective and prospective study, human salivary glands were investigated after radiation treatment for head and neck cancers. The aim was to assess acinar cell loss and morphologic changes after radiation therapy and to determine whether irradiated salivary glands have regenerative potential. METHODS AND MATERIALS Irradiated human submandibular and parotid salivary glands were collected from 16 patients at a range of time intervals after completion of radiation therapy (RT). Control samples were collected from 14 patients who had not received radiation treatments. Tissue sections were analyzed using immunohistochemistry to stain for molecular markers. RESULTS Human submandibular and parotid glands isolated less than 1 year after RT showed a near complete loss of acinar cells. However, acinar units expressing functional secretory markers were observed in all samples isolated at later intervals after RT. Significantly lower acinar cell numbers and increased fibrosis were found in glands treated with combined radiation and chemotherapy, in comparison to glands treated with RT alone. Irradiated samples showed increased staining for duct cell keratin markers, as well as many cells coexpressing acinar- and duct cell-specific markers, in comparison to nonirradiated control samples. CONCLUSIONS After RT, acinar cell clusters are maintained in human submandibular glands for years. The surviving acinar cells retain proliferative potential, although significant regeneration does not occur. Persistent DNA damage, increased fibrosis, and altered cell identity suggest mechanisms that may impair regeneration.
Collapse
Affiliation(s)
- Martha E Luitje
- Department of Otolaryngology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Anna-Karoline Israel
- Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Michael A Cummings
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Ellen J Giampoli
- Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Paul D Allen
- Department of Otolaryngology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Shawn D Newlands
- Department of Otolaryngology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Catherine E Ovitt
- Center for Oral Biology, Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
24
|
Witjes MJH, Vissink A. Cooperation as core value in an innovative era. Oral Dis 2020; 27:3-6. [PMID: 33006822 DOI: 10.1111/odi.13660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Max J H Witjes
- Department of Oral and Maxillofacial Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
25
|
Doppalapudi R, Vundavalli S, Prabhat MP. Effect of probiotic bacteria on oral Candida in head- and neck-radiotherapy patients: A randomized clinical trial. J Cancer Res Ther 2020; 16:470-477. [PMID: 32719253 DOI: 10.4103/jcrt.jcrt_334_18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objective The aim of the study is to assess the effect of probiotic bacteria on oral Candida counts in cancer patients who are undergoing head- and neck-radiotherapy in a tertiary care center. Study Design The study was a randomized clinical trial including 90 patients who just completed head- and neck-radiotherapy. Materials and Methods Participants were randomly allocated into three equal sized groups, i.e., probiotics group, candid group, and combination groups. Oral rinse samples of the patients were collected before and after the intervention for the identification of Candida. The samples were incubated on Sabouraud's Dextrose Agar with Chloramphenicol at 37°C for 48 h, to assess the counts of colony-forming units/milliliter (CFU/ml) of Candida in saliva, and further on chrome agar plates to identify the Candida spp. Data were analyzed using mixed ANOVA to compare mean CFU/ml of Candida among three groups before and after the intervention. Results A total of 86 patients were included in the final analysis and there was a statistically significant reduction in mean Candida spp. Counts (CFU/ml) after intervention in all the three groups (P = 0.000) and significant reductions identified in both probiotic and combination therapy groups. Apart from reduction in Candida albicans, significant decrease in Candida glabrata and Candida tropicalis was observed after probiotics usage compared to other groups. Conclusions The present study suggests that probiotic bacteria were effective in reducing oral Candida spp which can be recommended alone or in combination with traditional antifungal agents for effective reduction in oral Candida in head- and neck-radiotherapy patients.
Collapse
Affiliation(s)
- Radhika Doppalapudi
- Department of Oral Radiology, College of Dentistry, Aljouf University, Sakaka, Kingdom of Saudi Arabia
| | - Sudhakar Vundavalli
- Department of Preventive Dentistry, College of Dentistry, Aljouf University, Sakaka, Kingdom of Saudi Arabia
| | - M P Prabhat
- Department of Oral Medicine and Radiology, Drs. S and NR Siddhartha Institute of Dental Sciences, Gannavaram, Andhra Pradesh, India
| |
Collapse
|
26
|
Jensen SB, Vissink A, Limesand KH, Reyland ME. Salivary Gland Hypofunction and Xerostomia in Head and Neck Radiation Patients. J Natl Cancer Inst Monogr 2020; 2019:5551361. [PMID: 31425600 DOI: 10.1093/jncimonographs/lgz016] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The most manifest long-term consequences of radiation therapy in the head and neck cancer patient are salivary gland hypofunction and a sensation of oral dryness (xerostomia). METHODS This critical review addresses the consequences of radiation injury to salivary gland tissue, the clinical management of salivary gland hypofunction and xerostomia, and current and potential strategies to prevent or reduce radiation injury to salivary gland tissue or restore the function of radiation-injured salivary gland tissue. RESULTS Salivary gland hypofunction and xerostomia have severe implications for oral functioning, maintenance of oral and general health, and quality of life. Significant progress has been made to spare salivary gland function chiefly due to advances in radiation techniques. Other strategies have also been developed, e.g., radioprotectors, identification and preservation/expansion of salivary stem cells by stimulation with cholinergic muscarinic agonists, and application of new lubricating or stimulatory agents, surgical transfer of submandibular glands, and acupuncture. CONCLUSION Many advances to manage salivary gland hypofunction and xerostomia induced by radiation therapy still only offer partial protection since they are often of short duration, lack the protective effects of saliva, or potentially have significant adverse effects. Intensity-modulated radiation therapy (IMRT), and its next step, proton therapy, have the greatest potential as a management strategy for permanently preserving salivary gland function in head and neck cancer patients.Presently, gene transfer to supplement fluid formation and stem cell transfer to increase the regenerative potential in radiation-damaged salivary glands are promising approaches for regaining function and/or regeneration of radiation-damaged salivary gland tissue.
Collapse
Affiliation(s)
- Siri Beier Jensen
- Department of Dentistry and Oral Health, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center, Groningen, The Netherlands
| | | | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
27
|
Serrano Martinez P, Cinat D, van Luijk P, Baanstra M, de Haan G, Pringle S, Coppes RP. Mouse parotid salivary gland organoids for the in vitro study of stem cell radiation response. Oral Dis 2020; 27:52-63. [PMID: 32531849 PMCID: PMC7818507 DOI: 10.1111/odi.13475] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/18/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Hyposalivation-related xerostomia is an irreversible, untreatable, and frequent condition after radiotherapy for head and neck cancer. Stem cell therapy is an attractive option of treatment, but demands knowledge of stem cell functioning. Therefore, we aimed to develop a murine parotid gland organoid model to explore radiation response of stem cells in vitro. MATERIALS AND METHODS Single cells derived from murine parotid gland organoids were passaged in Matrigel with defined medium to assess self-renewal and differentiation potential. Single cells were irradiated and plated in a 3D clonogenic stem cell survival assay to assess submandibular and parotid gland radiation response. RESULTS Single cells derived from parotid gland organoids were able to extensively self-renew and differentiate into all major tissue cell types, indicating the presence of potential stem cells. FACS selection for known salivary gland stem cell markers CD24/CD29 did not further enrich for stem cells. The parotid gland organoid-derived stem cells displayed radiation dose-response curves similar to the submandibular gland. CONCLUSIONS Murine parotid gland organoids harbor stem cells with long-term expansion and differentiation potential. This model is useful for mechanistic studies of stem cell radiation response and suggests similar radiosensitivity for the parotid and submandibular gland organoids.
Collapse
Affiliation(s)
- Paola Serrano Martinez
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Davide Cinat
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter van Luijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Mirjam Baanstra
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerald de Haan
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robert P Coppes
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Spijkervet FKL, Schuurhuis JM, Stokman MA, Witjes MJH, Vissink A. Should oral foci of infection be removed before the onset of radiotherapy or chemotherapy? Oral Dis 2020; 27:7-13. [PMID: 32166855 PMCID: PMC7818265 DOI: 10.1111/odi.13329] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/19/2020] [Accepted: 03/05/2020] [Indexed: 01/22/2023]
Abstract
Pretreatment dental screening aims to locate and eliminate oral foci of infection in order to eliminate local, loco-regional, or systemic complications during and after oncologic treatment. An oral focus of infection is a pathologic process in the oral cavity that does not cause major infectious problems in healthy individuals, but may lead to severe local or systemic inflammation in patients subjected to oncologic treatment. As head and neck radiotherapy patients bear a lifelong risk on oral sequelae resulting from this therapy, the effects of chemotherapy on healthy oral tissues are essentially temporary and reversible. This has a large impact on what to consider as an oral focus of infection when patients are subjected to, for example, head and neck radiotherapy for cancer or intensive chemotherapy for hematological disorders. While in patients subjected to head and neck radiotherapy oral foci of infection have to be removed before therapy that may cause problems ultimately, in patients that will receive chemotherapy such, so-called chronic, foci of infection are not in need of removal of teeth but can be treated during a remission phase. Acute foci of infection always have to be removed before or early after the onset of any oncologic treatment.
Collapse
Affiliation(s)
- Fred K L Spijkervet
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jennifer M Schuurhuis
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Monique A Stokman
- Department of Radiotherapy, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Max J H Witjes
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Lim SB, Tsai CJ, Yu Y, Greer P, Fuangrod T, Hwang K, Fontenla S, Coffman F, Lee N, Lovelock DM. Investigation of a Novel Decision Support Metric for Head and Neck Adaptive Radiation Therapy Using a Real-Time In Vivo Portal Dosimetry System. Technol Cancer Res Treat 2020; 18:1533033819873629. [PMID: 31551011 PMCID: PMC6763934 DOI: 10.1177/1533033819873629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In adaptive radiation therapy of head and neck cancer, any significant anatomical changes observed are used to adapt the treatment plan to maintain target coverage without elevating the risk of xerostomia. However, the additional resources required for adaptive radiation therapy pose a challenge for broad-based implementation. It is hypothesized that a change in transit fluence is associated with volumetric change in the vicinity of the target and therefore can be used as a decision support metric for adaptive radiation therapy. This was evaluated by comparing the fluence with volumetric changes in 12 patients. Transit fluence was measured by an in vivo portal dosimetry system. Weekly cone beam computed tomography was used to determine volume change in the rectangular region of interest from condyloid process to C6. The integrated transit fluence through the region of interest on the day of the cone beam computed tomography scan was calculated with the first treatment as the baseline. The correlation between fluence change and volume change was determined. A logistic regression model was also used to associate the 5% region of interest volume reduction replanning trigger point and the fluence change. The model was assessed by a chi-square test. The area under the receiver-operating characteristic curve was also determined. A total of 46 pairs of measurements were obtained. The correlation between fluence and volumetric changes was found to be -0.776 (P value <.001). The negative correlation is attributed to the increase in the photon fluence transport resulting from the volume reduction. The chi-square of the logistic regression was found to be 17.4 (P value <.001). The area under the receiver-operating characteristic curve was found to be 0.88. Results indicate the change in transit fluence, which can be measured without consuming clinical resources or requiring additional time in the treatment room, can be used as a decision support metric for adaptive therapy.
Collapse
Affiliation(s)
- S B Lim
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Health Informatics, Rutgers University, Newark, NJ, USA
| | - C J Tsai
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Y Yu
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - P Greer
- Calvary Mater Newcastle Hospital, Newcastle, Australia
| | - T Fuangrod
- HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, BKK, Thailand
| | - K Hwang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Fontenla
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - F Coffman
- Department of Health Informatics, Rutgers University, Newark, NJ, USA
| | - N Lee
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - D M Lovelock
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
30
|
Spijkervet FKL, Brennan MT, Peterson DE, Witjes MJH, Vissink A. Research Frontiers in Oral Toxicities of Cancer Therapies: Osteoradionecrosis of the Jaws. J Natl Cancer Inst Monogr 2019; 2019:5551359. [DOI: 10.1093/jncimonographs/lgz006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/13/2019] [Indexed: 01/28/2023] Open
Abstract
AbstractThe deleterious effects of head and neck radiation on bone, with osteoradionecrosis (ORN) as the major disabling side effect of head and neck cancer treatment, are difficult to prevent and hard to treat. This review focuses on the current state of the science regarding the pathobiology, clinical impact, and management of ORN. With regard to the pathobiology underlying ORN, it is not yet confirmed whether the current radiation schedules by 3-dimensional conformal radiotherapy and intensity modified radiotherapy result in an unchanged, decreased, or increased risk of developing ORN when compared with conventional radiation treatment, the main risk factor being the total radiation dose delivered on any clinically significant surface of the mandible.With regard to the prevention of ORN, a thorough, early pre-irradiation dental assessment is still considered the first step to reduce the hazard of developing ORN post-radiotherapy, and hyperbaric oxygen (HBO) treatment reduces the risk of developing ORN in case of dental surgery in an irradiated field.With regard to the treatment of ORN, the focus is bidirectional: elimination of the necrotic bone and improving the vascularity of the normal tissues that were included in the radiation portal. The cure rate of limited ORN by conservative therapy is approximately 50%, and the cure rate of surgical approaches when conservative therapy has failed is approximately 40%.Whether it is effective to support conservative or surgical treatment with HBO as an adjuvant is not set. HBO treatment is shown to increase the vascularity of hard and soft tissues and has been reported to be beneficial in selected cases. However, in randomized clinical trials comparing the preventive effect of HBO on developing ORN with, eg, antibiotic coverage in patients needing dental surgery, the preventive effect of HBO was not shown to surpass that of a more conservative approach.More recently, pharmacologic management was introduced in the treatment of ORN with success, but its efficacy has to be confirmed in randomized clinical trials. The major problem of performing well-designed randomized clinical trials in ORN is having access to large numbers of patients with well-defined, comparable cases of ORN. Because many institutions will not have large numbers of such ORN cases, national and international scientific societies must be approached to join multicenter trials. Fortunately, the interest of funding organizations and the number researchers with an interest in healthy aging is growing. Research aimed at prevention and reduction of the morbidity of cancer treatment fits well within these programs.
Collapse
Affiliation(s)
- Frederik K L Spijkervet
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michael T Brennan
- Department of Oral Medicine, Carolinas Medical Center, Charlotte, NC
| | - Douglas E Peterson
- Department of Oral Health and Diagnostic Sciences, School of Dental Medicine, Neag Comprehensive Cancer Center, UConn Health, Farmington, CT
| | - Max J H Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Liu Y, Shi H, Huang S, Chen X, Zhou H, Chang H, Xia Y, Wang G, Yang X. Early prediction of acute xerostomia during radiation therapy for nasopharyngeal cancer based on delta radiomics from CT images. Quant Imaging Med Surg 2019; 9:1288-1302. [PMID: 31448214 DOI: 10.21037/qims.2019.07.08] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Acute xerostomia is the most common side effect of radiation therapy (RT) for head and neck (H&N) malignancies. Investigating radiation-induced changes of computed tomography (CT) radiomics in parotid glands (PGs) and saliva amount (SA) can predict acute xerostomia during the RT for nasopharyngeal cancer (NPC). Methods CT and SA data from 35 patients with stages I-IVB were randomly collected from an NPC clinical trial registered on the clinicaltrials.gov (ID: NCT01762514). All patients received radical treatment based on intensity-modulated RT (IMRT) with a prescription dose of 68.1 Gy in 30 fractions. The patients' ages ranged 24-72 years, and each patient had five CT sets acquired at treatment position: at the 0th, 10th, 20th, 30th fractions during the RT, and at 3-month later after the RT. The PGs for each CT set were delineated by a radiation oncologist and verified independently by another. Patients' saliva was collected every other 10 days during the RT. Acute xerostomia was evaluated based on the RTOG acute toxicity scoring and the SA. In total, 1,703 radiomics features were calculated for PGs from each CT set, including feature value at 0th fraction (FV0F), FV10F, and delta FV (ΔFV10F-0F), respectively. Extensive experiments were conducted to achieve the optimal results. RidgeCV and Recursive Feature Elimination (RFE) were used for feature selection, while linear regression was used for predicting SA30F. Four more patients were added for independent testing. Results Substantial changes in various radiomics metrics of PGs were observed during the RT. Eight normalized feature value (NFV), selected from NFV0F, predicted SA10F with a mean square error (MSE) of 0.9042 and a R2 score of 0.7406. Fourteen NFV, selected from ΔNFV10F-0F, NFV0F, and NFV10F to predict SA30F, showed the best predictive ability with an MSE of 0.0569. The model predicted the level of acute xerostomia with a precision of 0.9220 and a sensitivity of 100%, compared to the clinical observed SA. For the independent test, the MSE of PSA30F was 0.0233. Conclusions This study demonstrated that radiation-induced acute xerostomia level could be early predicted based on the SA and radiomics changes of the PGs during IMRT delivery. SA, NFV0F, NFV10F, and especially ΔNFV10F-0F provided the best performance on acute xerostomia prediction for individual patient based on RidgeCV_RFE_LinearRegression method of delta radiomics.
Collapse
Affiliation(s)
- Yanxia Liu
- School of Software Engineering, South China University of Technology, Guangzhou 510006, China
| | - Hongyu Shi
- School of Software Engineering, South China University of Technology, Guangzhou 510006, China
| | - Sijuan Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
| | - Xiaochuan Chen
- School of Software Engineering, South China University of Technology, Guangzhou 510006, China
| | - Huimin Zhou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China.,Department of Oncology, the Seventy-fourth Group Army Hospital of the Chinese People's Liberation Army, Guangzhou 510318, China
| | - Hui Chang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
| | - Yunfei Xia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
| | - Guohua Wang
- School of Software Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xin Yang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
| |
Collapse
|
32
|
Men K, Geng H, Zhong H, Fan Y, Lin A, Xiao Y. A Deep Learning Model for Predicting Xerostomia Due to Radiation Therapy for Head and Neck Squamous Cell Carcinoma in the RTOG 0522 Clinical Trial. Int J Radiat Oncol Biol Phys 2019; 105:440-447. [PMID: 31201897 DOI: 10.1016/j.ijrobp.2019.06.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/14/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE Xerostomia commonly occurs in patients who undergo head and neck radiation therapy and can seriously affect patients' quality of life. In this study, we developed a xerostomia prediction model with radiation treatment data using a 3-dimensional (3D) residual convolutional neural network (rCNN). The model can be used to guide radiation therapy to reduce toxicity. METHODS AND MATERIALS A total of 784 patients with head and neck squamous cell carcinoma enrolled in the Radiation Therapy Oncology Group 0522 clinical trial were included in this study. Late xerostomia is defined as xerostomia of grade ≥2 occurring in the 12th month of radiation therapy. The computed tomography (CT) planning images, 3D dose distributions, and contours of the parotid and submandibular glands were included as 3D rCNN inputs. Comparative experiments were performed for the 3D rCNN model without 1 of the 3 inputs and for the logistic regression model. Accuracy, sensitivity, specificity, F-score, and area under the receiver operator characteristic curve were evaluated. RESULTS The proposed model achieved promising prediction results. The performance metrics for 3D rCNN model with contour, CT images, and radiation therapy dose; 3D rCNN without contour; 3D rCNN without CT images; 3D rCNN without the dose; logistic regression with the dose and clinical parameters; and logistic regression without clinical parameters were as follows: accuracy: 0.76, 0.74, 0.73, 0.65, 0.64, and 0.56; sensitivity: 0.76, 0.72, 0.77, 0.59, 0.72, and 0.75; specificity: 0.76, 0.76, 0.71, 0.69, 0.59, and 0.43; F-score: 0.70, 0.68, 0.69, 0.56, 0.60, and 0.57; and area under the receiver operator characteristic curve: 0.84, 0.82, 0.78, 0.70, 0.74, and 0.68, respectively. CONCLUSIONS The proposed model uses 3D rCNN filters to extract low- and high-level spatial features and to achieve promising performance. This is a potentially effective model for predicting objective toxicity for supporting clinical decision making.
Collapse
Affiliation(s)
- Kuo Men
- University of Pennsylvania, Philadelphia, Pennsylvania; National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Huaizhi Geng
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - Haoyu Zhong
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yong Fan
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Lin
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ying Xiao
- University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Urkasemsin G, Ferreira JN. Unveiling Stem Cell Heterogeneity Toward the Development of Salivary Gland Regenerative Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1123:151-164. [PMID: 31016599 DOI: 10.1007/978-3-030-11096-3_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epithelial damage in the salivary gland (SG) resulting in irreversible dry mouth can be commonly induced by gamma radiation therapy. This radiation depletes the SG stem/progenitor cell niche slowing healing and natural gland regeneration. Biologists have been focused in understanding the development and differentiation of epithelial stem and progenitor cell niches during SG organogenesis. These organogenesis studies gave insights into novel cell-based therapies to recreate the three-dimensional (3D) salivary gland (SG) organ, recapitulate the SG native physiology, and restore saliva secretion. Such therapeutical strategies apply techniques that assemble, in a 3D organotypic culture, progenitor and stem cell lines to develop SG organ-like organoids or mini-transplants. Future studies will employ a combination of organoids, decellularized matrices, and smart biomaterials to create viable and functional SG transplants to repair the site of SG injury and reestablish saliva production.
Collapse
Affiliation(s)
- Ganokon Urkasemsin
- Faculty of Veterinary Science, Department of Preclinical and Applied Animal Science, Mahidol University, Nakhon Pathom, Thailand
| | - Joao N Ferreira
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand. .,National University of Singapore, Singapore, Singapore.
| |
Collapse
|
34
|
Zhang XM, Huang Y, Cong X, Qu LH, Zhang K, Wu LL, Zhang Y, Yu GY. Parasympathectomy increases resting secretion of the submandibular gland in minipigs in the long term. J Cell Physiol 2018; 234:9515-9524. [PMID: 30387129 DOI: 10.1002/jcp.27640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/02/2018] [Indexed: 11/09/2022]
Abstract
Parasympathectomy leads to retrogressive alteration and dysfunction of the submandibular gland (SMG) within 1 month, but its long-term effect is unclear. Excessive secretion is observed in half of the patients 4-6 months after SMG transplantation, which completely denervates the gland. Here, we investigated the long-term effect of parasympathectomy on the secretion of SMGs in minipigs. The results showed that the resting salivary secretion of SMGs decreased by 82.9% of that in control at 2 months after denervation, but increased by 156% at 6 months. Although experiencing an atrophic period, the denervated glands regained their normal morphology by 6 months. The expression of the function-related proteins, including muscarinic acetylcholine receptor (mAChR) 3, aquaporin 5 (AQP5), tight junction protein claudin-3, and claudin-4 was decreased at 2 months after denervation. Meanwhile, the protein expression of stem cell markers, including sex-determining region Y-box 2 and octamer-binding transcription factor 4, and the number of Ki67+ cells were significantly increased. However, at 6 months after denervation, the expression of mAChR3, AQP5, claudin-1, claudin-3, and claudin-4 was significantly raised, and the membrane distribution of these proteins was increased accordingly. The autonomic axonal area of the glands was reduced at 2 months after denervation but returned to the control level at 6 months, suggesting that reinnervation took place in the long term. In summary, parasympathectomy increases resting secretion of the SMGs in the long term with a possible mechanism involving improved transepithelial fluid transport. This finding may provide a new strategy for xerostomia treatment.
Collapse
Affiliation(s)
- Xue-Ming Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan Huang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Ling-Han Qu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Kuo Zhang
- Department of Laboratory Animal Science, Peking University Health Science Center, Beijing, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
35
|
Involvement of Aquaporins in the Pathogenesis, Diagnosis and Treatment of Sjögren's Syndrome. Int J Mol Sci 2018; 19:ijms19113392. [PMID: 30380700 PMCID: PMC6274940 DOI: 10.3390/ijms19113392] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/24/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023] Open
Abstract
Sjögren’s syndrome (SS) is a chronic autoimmune disease characterized by lymphocytic infiltration of salivary and lacrimal glands resulting in diminished production of saliva and tears. The pathophysiology of SS has not yet been fully deciphered. Classically it has been postulated that sicca symptoms in SS patients are a double step process whereby lymphocytic infiltration of lacrimal and salivary glands (SG) is followed by epithelial cell destruction resulting in keratoconjunctivitis sicca and xerostomia. Recent advances in the field of the pathophysiology of SS have brought in new players, such as aquaporins (AQPs) and anti AQPs autoantibodies that could explain underlying mechanistic processes and unveil new pathophysiological pathways offering a deeper understanding of the disease. In this review, we delineate the link between the AQP and SS, focusing on salivary glands, and discuss the role of AQPs in the treatment of SS-induced xerostomia.
Collapse
|
36
|
Wong WY, Pier M, Limesand KH. Persistent disruption of lateral junctional complexes and actin cytoskeleton in parotid salivary glands following radiation treatment. Am J Physiol Regul Integr Comp Physiol 2018; 315:R656-R667. [PMID: 29897817 PMCID: PMC6230885 DOI: 10.1152/ajpregu.00388.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 02/02/2023]
Abstract
Xerostomia and hyposalivation are debilitating side effects for patients treated with ionizing radiation for head and neck cancer. Despite technological advances, collateral damage to the salivary glands remains a significant problem for patients and severely diminishes their quality of life. During the wound healing process, restoration of junctional contacts is necessary to maintain polarity, structural integrity, and orientation cues for secretion. However, little is known about whether these structural molecules are impacted following radiation damage and more importantly, during tissue restoration. We evaluated changes in adherens junctions and cytoskeletal regulators in an injury model where mice were irradiated with 5 Gy and a restoration model where mice injected postradiation with insulin-like growth factor 1 (IGF1) are capable of restoring salivary function. Using coimmunoprecipitation, there is a decrease in epithelial (E)-cadherin bound to β-catenin following damage that is restored to untreated levels with IGF1. Via its adaptor proteins, β-catenin links the cadherins to the cytoskeleton and part of this regulation is mediated through Rho-associated coiled-coil containing kinase (ROCK) signaling. In our radiation model, filamentous (F)-actin organization is fragmented, and there is an induction of ROCK activity. However, a ROCK inhibitor, Y-27632, prevents E-cadherin/β-catenin dissociation following radiation treatment. These findings illustrate that radiation induces a ROCK-dependent disruption of the cadherin-catenin complex and alters F-actin organization at stages of damage when hyposalivation is observed. Understanding the regulation of these components will be critical in the discovery of therapeutics that have the potential to restore function in polarized epithelium.
Collapse
Affiliation(s)
- Wen Yu Wong
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona , Tucson, Arizona
| | - Maricela Pier
- Department of Nutritional Sciences, University of Arizona , Tucson, Arizona
| | - Kirsten H Limesand
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona , Tucson, Arizona
- Department of Nutritional Sciences, University of Arizona , Tucson, Arizona
| |
Collapse
|
37
|
Kwak M, Ninche N, Klein S, Saur D, Ghazizadeh S. c-Kit + Cells in Adult Salivary Glands do not Function as Tissue Stem Cells. Sci Rep 2018; 8:14193. [PMID: 30242278 PMCID: PMC6155036 DOI: 10.1038/s41598-018-32557-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/10/2018] [Indexed: 11/01/2022] Open
Abstract
A rare population of salivary gland cells isolated based on c-Kit immunoreactivity are thought to represent tissue stem cells since they exhibit the most robust proliferative and differentiation capacity ex vivo. Despite their high promise for cell-based therapies aimed at restoring salivary function, the precise location and in vivo function of c-Kit+ stem cells remain unclear. Here, by combining immunostaining with c-KitCreERT2-based genetic labeling and lineage tracing in the adult mouse salivary glands, we show that c-Kit is expressed in a relatively large and heterogeneous cell population that consists mostly of differentiated cells. Moreover, c-Kit does not mark ductal stem cells that are known to express cytokeratin K14. Tracking the fate of in vivo-labeled c-Kit+ or that of K14+ cells in spheroid cultures reveals a limited proliferative potential for c-Kit+ cells and identifies K14+ cells as the major source of salispheres in these cultures. Long-term in vivo lineage tracing studies indicate that although c-Kit marks at least two discrete ductal cell lineages, c-Kit+ cells do not contribute to the normal maintenance of any other cell lineages. Our results indicate that c-Kit is not a reliable marker for salivary gland stem cells, which has important implications for salivary gland regenerative therapies.
Collapse
Affiliation(s)
- Mingyu Kwak
- Department of Oral Biology & Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Ninche Ninche
- Department of Oral Biology & Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sabine Klein
- Department of Internal Medicine, Technical University of Munich, München, Germany
| | - Dieter Saur
- Department of Internal Medicine, Technical University of Munich, München, Germany
| | - Soosan Ghazizadeh
- Department of Oral Biology & Pathology, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
38
|
Nagle PW, Hosper NA, Barazzuol L, Jellema AL, Baanstra M, van Goethem MJ, Brandenburg S, Giesen U, Langendijk JA, van Luijk P, Coppes RP. Lack of DNA Damage Response at Low Radiation Doses in Adult Stem Cells Contributes to Organ Dysfunction. Clin Cancer Res 2018; 24:6583-6593. [PMID: 30135147 DOI: 10.1158/1078-0432.ccr-18-0533] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/08/2018] [Accepted: 08/17/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Radiotherapy for head and neck cancer may result in serious side effects, such as hyposalivation, impairing the patient's quality of life. Modern radiotherapy techniques attempt to reduce the dose to salivary glands, which, however, results in low-dose irradiation of the tissue stem cells. Here we assess the low-dose sensitivity of tissue stem cells and the consequences for tissue function. EXPERIMENTAL DESIGN Postirradiation rat salivary gland secretory function was determined after pilocarpine induction. Murine and patient-derived salivary gland and thyroid gland organoids were irradiated and clonogenic survival was assessed. The DNA damage response (DDR) was analyzed in organoids and modulated using different radiation modalities, chemical inhibition, and genetic modification. RESULTS Relative low-dose irradiation to the high-density stem cell region of rat salivary gland disproportionally impaired function. Hyper-radiosensitivity at doses <1 Gy, followed by relative radioresistance at doses ≥1 Gy, was observed in salivary gland and thyroid gland organoid cultures. DDR modulation resulted in diminished, or even abrogated, relative radioresistance. Furthermore, inhibition of the DDR protein ATM impaired DNA repair after 1 Gy, but not 0.25 Gy. Irradiation of patient-derived salivary gland organoid cells showed similar responses, whereas a single 1 Gy dose to salivary gland-derived stem cells resulted in greater survival than clinically relevant fractionated doses of 4 × 0.25 Gy. CONCLUSIONS We show that murine and human glandular tissue stem cells exhibit a dose threshold in DDR activation, resulting in low-dose hyper-radiosensitivity, with clinical implications in radiotherapy treatment planning. Furthermore, our results from patient-derived organoids highlight the potential of organoids to study normal tissue responses to radiation.
Collapse
Affiliation(s)
- Peter W Nagle
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nynke A Hosper
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne L Jellema
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mirjam Baanstra
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marc-Jan van Goethem
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,KVI Center for Advanced Radiation Technology, University of Groningen, Groningen, the Netherlands
| | - Sytze Brandenburg
- KVI Center for Advanced Radiation Technology, University of Groningen, Groningen, the Netherlands
| | - Ulrich Giesen
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig, Germany
| | - Johannes A Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter van Luijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rob P Coppes
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands. .,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
39
|
|
40
|
Assy Z, Brand HS. A systematic review of the effects of acupuncture on xerostomia and hyposalivation. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:57. [PMID: 29439690 PMCID: PMC5811978 DOI: 10.1186/s12906-018-2124-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 02/05/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Saliva is fundamental to our oral health and our well-being. Many factors can impair saliva secretion, such as adverse effects of prescribed medication, auto-immune diseases (for example Sjögren's syndrome) and radiotherapy for head and neck cancers. Several studies have suggested a positive effect of acupuncture on oral dryness. METHODS Pubmed and Web of Science were electronically searched. Reference lists of the included studies and relevant reviews were manually searched. Studies that met the inclusion criteria were systematically evaluated. Two reviewers assessed each of the included studies to confirm eligibility and assessing the risk of bias. RESULTS Ten randomized controlled trials investigating the effect of acupuncture were included. Five trials compared acupuncture to sham/placebo acupuncture. Four trials compared acupuncture to oral hygiene/usual care. Only one clinical trial used oral care sessions as control group. For all the included studies, the quality for all the main outcomes has been assessed as low. Although some publications suggest a positive effect of acupuncture on either salivary flow rate or subjective dry mouth feeling, the studies are inconclusive about the potential effects of acupuncture. CONCLUSIONS Insufficient evidence is available to conclude whether acupuncture is an evidence-based treatment option for xerostomia/hyposalivation. Further well-designed, larger, double blinded trials are required to determine the potential benefit of acupuncture. Sample size calculations should be performed before before initiating these studies.
Collapse
Affiliation(s)
- Zainab Assy
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), room 12N-37, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Henk S. Brand
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), room 12N-37, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| |
Collapse
|
41
|
Malallah OS, Garcia CMA, Proctor GB, Forbes B, Royall PG. Buccal drug delivery technologies for patient-centred treatment of radiation-induced xerostomia (dry mouth). Int J Pharm 2018; 541:157-166. [PMID: 29425763 DOI: 10.1016/j.ijpharm.2018.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/29/2018] [Accepted: 02/04/2018] [Indexed: 01/01/2023]
Abstract
Radiotherapy is a life-saving treatment for head and neck cancers, but almost 100% of patients develop dry mouth (xerostomia) because of radiation-induced damage to their salivary glands. Patients with xerostomia suffer symptoms that severely affect their health as well as physical, social and emotional aspects of their life. The current management of xerostomia is the application of saliva substitutes or systemic delivery of saliva-stimulating cholinergic agents, including pilocarpine, cevimeline or bethanechol tablets. It is almost impossible for substitutes to replicate all the functional and sensory facets of natural saliva. Salivary stimulants are a better treatment option than saliva substitutes as the former induce the secretion of natural saliva from undamaged glands; typically, these are the minor salivary glands. However, patients taking cholinergic agents systemically experience pharmacology-related side effects including sweating, excessive lacrimation and gastrointestinal tract distresses. Local delivery direct to the buccal mucosa has the potential to provide rapid onset of drug action, i.e. activation of minor salivary glands within the buccal mucosa, while sparing systemic drug exposure and off-target effects. This critical review of the technologies for the local delivery of saliva-stimulating agents includes oral disintegrating tablets (ODTs), oral disintegrating films, medicated chewing gums and implantable drug delivery devices. Our analysis makes a strong case for the development of ODTs for the buccal delivery of cholinergic agents: these must be patient-friendly delivery platforms with variable loading capacities that release the drug rapidly in fluid volumes typical of residual saliva in xerostomia (0.05-0.1 mL).
Collapse
Affiliation(s)
- Osamah S Malallah
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, UK
| | | | - Gordon B Proctor
- Institute of Mucosal & Salivary biology, King's College London, Thomas Street, London SE1 9RT, UK
| | - Ben Forbes
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, UK
| | - Paul G Royall
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
42
|
Hawkins PG, Kadam AS, Jackson WC, Eisbruch A. Organ-Sparing in Radiotherapy for Head-and-Neck Cancer: Improving Quality of Life. Semin Radiat Oncol 2018; 28:46-52. [DOI: 10.1016/j.semradonc.2017.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Sakai Y, Yamamori T, Yoshikawa Y, Bo T, Suzuki M, Yamamoto K, Ago T, Inanami O. NADPH oxidase 4 mediates ROS production in radiation-induced senescent cells and promotes migration of inflammatory cells. Free Radic Res 2017; 52:92-102. [PMID: 29228832 DOI: 10.1080/10715762.2017.1416112] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Excessive DNA damage induced by ionising radiation (IR) to normal tissue cells is known to trigger cellular senescence, a process termed stress-induced premature senescence (SIPS). SIPS is often accompanied by the production of reactive oxygen species (ROS), and this is reported to be important for the initiation and maintenance of SIPS. However, the source of ROS during SIPS after IR and their significance in radiation-induced normal tissue damage remain elusive. In the present study, we tested the hypothesis that the NADPH oxidase (NOX) family of proteins mediates ROS production in SIPS-induced cells after IR and plays a role in SIPS-associated biological events. X-irradiation of primary mouse embryonic fibroblasts (MEFs) resulted in cellular senescence and the concomitant increase of intracellular ROS. Among all six murine NOX isoforms (NOX1-4 and DUOX1/2), only NOX4 was detectable under basal conditions and was upregulated following IR. In addition, radiation-induced ROS production was diminished by genetic or pharmacological inhibition of NOX4. Meanwhile, NOX4 deficiency did not affect the induction of cellular senescence after IR. Furthermore, the migration of human monocytic U937 cells to the culture medium collected from irradiated MEFs was significantly reduced by NOX4 inhibition, suggesting that NOX4 promotes the recruitment of inflammatory cells. Collectively, our findings imply that NOX4 mediates ROS production in radiation-induced senescent cells and contributes to normal tissue damage after IR via the recruitment of inflammatory cells and the exacerbation of tissue inflammation.
Collapse
Affiliation(s)
- Yuri Sakai
- a Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Tohru Yamamori
- a Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Yoji Yoshikawa
- b Department of Medicine and Clinical Science, Graduate School of Medical Sciences , Kyushu University , Fukuoka , Japan
| | - Tomoki Bo
- a Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Motofumi Suzuki
- c Radiation and Cancer Biology Team , National Institutes for Quantum and Radiobiological Science and Technology , Chiba , Japan
| | - Kumiko Yamamoto
- a Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Tetsuro Ago
- b Department of Medicine and Clinical Science, Graduate School of Medical Sciences , Kyushu University , Fukuoka , Japan
| | - Osamu Inanami
- a Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| |
Collapse
|
44
|
CRISPR-Cas9 HDR system enhances AQP1 gene expression. Oncotarget 2017; 8:111683-111696. [PMID: 29340084 PMCID: PMC5762352 DOI: 10.18632/oncotarget.22901] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 11/16/2017] [Indexed: 01/04/2023] Open
Abstract
Ionizing radiation (IR) isthe primarytherapeutic tool to treat patients with cancerous lesions located in the head and neck. In many patients, IR results in irreversible and severe salivary gland dysfunction or xerostomia. Currently there are no effective treatment options to reduce the effects of xerostomia. More recently, salivary gland gene therapy utilizing the water-specific protein aquaporin 1 (AQP1) has been of great interest to potentially correct salivary dysfunction. In this study, we used CRISPR-Cas9 gene editing along with the endogenous promoter of AQP1 within theHEK293 and MDCK cell lines. The successful integration of the cytomegalovirus (CMV) promoterresultedin a significant increase of AQP1 gene transcription and translation. Additionalfunctional experiments involvingthe MDCK cell line confirmedthat over-expressed AQP1increasedtransmembrane fluid flux indicative of increased intracellular fluid flux. The off-target effect of designed guided RNA sequence was analyzed and demonstrateda high specificity for the Cas9 cleavage. Considering the development of new methods for robust DNA knock-in, our results suggest that endogenous promoter replacement may be a potential treatment forsalivary gland dysfunction.
Collapse
|
45
|
Fang D, Shang S, Liu Y, Bakkar M, Sumita Y, Seuntjens J, Tran SD. Optimal timing and frequency of bone marrow soup therapy for functional restoration of salivary glands injured by single-dose or fractionated irradiation. J Tissue Eng Regen Med 2017; 12:e1195-e1205. [PMID: 28714550 DOI: 10.1002/term.2513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 03/07/2017] [Accepted: 06/20/2017] [Indexed: 12/30/2022]
Abstract
Injections of bone marrow (BM) cell extract, known as 'BM soup', were previously reported to mitigate ionizing radiation (IR) injury to salivary glands (SGs). However, the optimal starting time and frequency to maintain BM soup therapeutic efficacy remains unknown. This study tested the optimal starting time and frequency of BM soup injections in mice radiated with either a single dose or a fractionated dose. First, BM soup treatment was started at 1, 3 or 7 weeks post-IR; positive (non-IR) and negative (IR) control mice received injections of saline (vehicle control). Second, BM soup-treated mice received injections at different frequencies (1, 2, 3 and 5 weekly injections). Third, a 'fractionated-dose radiation' model to injure mouse SGs was developed (5 Gy × 5 days) and compared with the single high dose radiation model. All mice (n = 65) were followed for 16 weeks post-IR. The results showed that starting injections of BM soup between 1 and 3 weeks mitigated the effect of IR-induced injury to SGs and improved the restoration of salivary function. Although the therapeutic effect of BM soup lessens after 8 weeks, it can be sustained by increasing the frequency of weekly injections. Moreover, both single-dose and fractionated-dose radiation models are efficient and comparable in inducing SG injury and BM soup treatments are effective in restoring salivary function in both radiation models. In conclusion, starting injections of BM soup within 3 weeks post-radiation, with 5 weekly injections, maintains 90-100% of saliva flow in radiated mice.
Collapse
Affiliation(s)
- Dongdong Fang
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Canada
| | - Sixia Shang
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Canada.,Department of Stomatology, People's Hospital of Dongying, Dongying, China
| | - Younan Liu
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Canada
| | - Mohammed Bakkar
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Canada
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Nagasaki University, Nagasaki, Japan
| | - Jan Seuntjens
- Department of Oncology, Medical Physics Unit, McGill University, Montreal, Canada
| | - Simon D Tran
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Canada
| |
Collapse
|
46
|
Metwalli KA, Do MA, Nguyen K, Mallick S, Kin K, Farokhnia N, Jun G, Fakhouri WD. Interferon Regulatory Factor 6 Is Necessary for Salivary Glands and Pancreas Development. J Dent Res 2017; 97:226-236. [PMID: 28898113 DOI: 10.1177/0022034517729803] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Interferon regulatory factor 6 ( IRF6) acts as a tumor suppressor and controls cell differentiation in ectodermal and craniofacial tissues by regulating expression of target genes. Haploinsufficiency of IRF6 causes Van der Woude and popliteal pterygium syndrome, 2 syndromic forms of cleft lip and palate. Around 85% of patients with Van der Woude express pits on the lower lip that continuously or intermittently drain saliva, and patients with the common cleft lip and palate have a higher prevalence of dental caries and gingivitis. This study aims to identify the role of IRF6 in development of exocrine glands, specifically the major salivary glands. Our transgenic mouse model that expresses LacZ reporter under the control of the human IRF6 enhancer element showed high expression of IRF6 in major and minor salivary glands and ducts. Immunostaining data also confirmed the endogenous expression of IRF6 in the developing ductal, serous, and mucous acinar cells of salivary glands. As such, we hypothesized that Irf6 is important for proper development of salivary glands and potentially other exocrine glands. Loss of Irf6 in mice causes an increase in the proliferation level of salivary cells, disorganized branching morphogenesis, and a lack of differentiated mucous acinar cells in submandibular and sublingual glands. Expression and localization of the acinar differentiation marker MIST1 were altered in Irf6-null salivary gland and pancreas. The RNA-Seq analysis demonstrated that 168 genes are differentially expressed and confer functions associated with transmembrane transporter activity, spliceosome, and transcriptional regulation. Furthermore, expression of genes involved in the EGF pathway-that is, Ereg, Ltbp4, Matn1, Matn3, and Tpo-was decreased at embryonic day 14.5, while levels of apoptotic proteins were elevated at postnatal day 0. In conclusion, our data report a novel role of Irf6 in exocrine gland development and support a rationale for performing exocrine functional tests for patients with IRF6-damaging mutations.
Collapse
Affiliation(s)
- K A Metwalli
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - M A Do
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - K Nguyen
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - S Mallick
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - K Kin
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - N Farokhnia
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - G Jun
- 2 Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - W D Fakhouri
- 1 Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA.,3 Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
47
|
Consumption and direct costs of dental care for patients with head and neck cancer: A 16-year cohort study. PLoS One 2017; 12:e0182877. [PMID: 28832673 PMCID: PMC5568378 DOI: 10.1371/journal.pone.0182877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/26/2017] [Indexed: 02/03/2023] Open
Abstract
Patients with head and neck (H&N) cancer are commonly treated with surgery and/or radiotherapy, which can increase the risk of oral infection, dental caries, and periodontal disease. The present study investigated dental care consumption and costs in patient with H&N cancer before and after the cancer diagnosis. Data from Swedish regional and national registers were used to follow up dental care utilization and dental procedure costs. The analysis included 2,754 patients who had been diagnosed with H&N cancer (exposed cohort) in Stockholm County, Sweden, during 2000–2012 and 13,036 matched persons without cancer (unexposed cohort). The exposed cohort was sub-grouped into irradiated and non-irradiated patients for analysis. The exposed cohort underwent a moderately higher number of dental procedures per year than the unexposed cohort in both the year of the cancer diagnosis and the year after cancer diagnosis; in addition, these numbers were higher in the irradiated than in the non-irradiated subgroup of the exposed cohort. Dental care consumption and costs in the exposed cohort declined over time but remained at a slightly higher level than in the unexposed cohort over the long term (more than two years). Examinations and preventive procedures accounted for most of the higher consumption in the short term (2 years) and at the longer term follow-up. Swedish national insurance subsidized costs for dental treatment, which were highest in the irradiated subgroup and lowest in the unexposed cohort. Direct costs to the patient, however, were similar among the groups. Swedish national health insurance protects patients with H&N cancer from high dental expenditures. Further studies on the cost-effectiveness of preventive dental care for patients are needed.
Collapse
|
48
|
Jaguar GC, Prado JD, Campanhã D, Alves FA. Clinical features and preventive therapies of radiation-induced xerostomia in head and neck cancer patient: a literature review. ACTA ACUST UNITED AC 2017. [DOI: 10.1186/s41241-017-0037-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
49
|
Choi JS, An HY, Shin HS, Kim YM, Lim JY. Enhanced tissue remodelling efficacy of adipose-derived mesenchymal stem cells using injectable matrices in radiation-damaged salivary gland model. J Tissue Eng Regen Med 2017; 12:e695-e706. [PMID: 27860388 DOI: 10.1002/term.2352] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/10/2016] [Accepted: 11/09/2016] [Indexed: 12/11/2022]
Abstract
The present study was conducted to introduce the use of a delivery carrier for local transplantation of human adipose tissue-derived mesenchymal stem cells (AdMSCs) into the salivary gland (SG) and analyse its ability to enhance radioprotection of AdMSCs against irradiation (IR)-induced damage. An injectable porcine small intestinal submucosa (SIS) matrix was used as a cell delivery carrier, and human AdMSCs were contained within SIS hydrogel (AdMSC/SIS). After local injection into SGs of mice following local IR, morphological and functional changes were evaluated in the sham, vehicle [phosphate-buffered saline (PBS)], SIS, AdMSC and AdMSC/SIS groups. Local transplantation of AdMSC resulted in less fibrosis, regardless of the use of a carrier, but the AdMSC/SIS group showed more mucin-producing acini relative to those in the PBS group. Functional restoration of salivation capacity and salivary protein synthesis was achieved in AdMSC and AdMSC/SIS groups, with a greater tendency being observed in the AdMSC/SIS group. AdMSC treatment resulted in tissue remodelling with a greater number of salivary epithelial cells (AQP-5), SG progenitor cells (c-Kit), endothelial cells (CD31) and myoepithelial cells (α-SMA), among which endothelial and myoepithelial cells significantly increased in the AdMSC/SIS group relative to the AdMSC group. AdMSC treatment alleviated IR-induced cell death, and the anti-apoptotic and anti-oxidative effects of AdMSC were enhanced in the AdMSC/SIS group relative to the AdMSC group. These results suggest local transplantation of AdMSC improves tissue remodelling following radiation damage in SG tissue, and that use of a carrier enhances the protective effects of AdMSC-mediated cellular protection against IR via paracrine secretion. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jeong-Seok Choi
- Department of Otorhinolaryngology - Head and Neck Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Hye-Young An
- Department of Otorhinolaryngology - Head and Neck Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Hyun-Soo Shin
- Department of Otorhinolaryngology - Head and Neck Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Young-Mo Kim
- Department of Otorhinolaryngology - Head and Neck Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology - Head and Neck Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| |
Collapse
|
50
|
Krishnan M, Tennavan A, Saraswathy S, Sekhri T, Singh AK, Nair V. Acute Radiation-Induced Changes in Sprague-Dawley Rat Submandibular Glands: A Histomorphometric Analysis. World J Oncol 2017; 8:45-52. [PMID: 29147434 PMCID: PMC5649996 DOI: 10.14740/wjon1021w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2017] [Indexed: 01/22/2023] Open
Abstract
Background Radiation-induced xerostomia is a distressing clinical condition that starts appearing from the initial stages of radiotherapy in head and neck cancer patients. Though submandibular glands contribute to maximum of the "resting salivary" secretions, most of the acute xerostomia experiments so far reported have been on animal parotid glands. Therefore, we assessed and quantified the histologic changes in submandibular glands of Sprague-Dawley (SD) rats using histomorphometry, 24 hours after radiation. Methods Three SD rats were given single-dose radiation of 15 Gray from a gamma cobalt-60 irradiator. Same number of non-radiated animals was the controls. Animals were sacrificed at 24 hours followed by histopathology and histomorphometry of submandibular glands, where the mean values were analyzed by Student's t-test. Results Irradiated submandibular glands showed highly significant reduction in acinar area (53%: 77.16±5.05% to 36.55±4.90%) and acinar size (87%: 3,447.53 ± 461.03 mm2 to 428.25 ± 75.22 mm2) with concomitant increase in inter-acinar space (3.46 ± 0.67 mm to 10.08 ± 0.60 mm). Acini nuclei displayed anisonucleosis, poikilonucleosis and pyknosis. "Serous acini" had marked morphologic changes, with fluid accumulation between cells, generalized cytoplasmic vacuolation and vascular congestion, while "mucous acini" largely retained cell architecture. Similarly, ductal cells and nuclei also did not show apparent differences. This demonstrated radiosensitivity variations among different submandibular gland cell types. Conclusion Evaluation of acute submandibular acinar cell dysfunctions has helped in quantifying the histologic elements, which mainly contribute to the resting salivary secretions. Findings would aid in future research of radioprotector drugs, salivary glandular regeneration modalities and in devising prudent radiotherapy protocols to address radiation-induced xerostomia.
Collapse
Affiliation(s)
- Manu Krishnan
- Department of Dental Research & Implantology, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Delhi, India
| | - Aatish Tennavan
- Department of Dental Research & Implantology, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Delhi, India
| | - Seema Saraswathy
- School of Medicine and Paramedical Health Sciences, Guru Gobind Singh Indraprastha University, Delhi, India
| | - Tarun Sekhri
- Division of Health, Endocrinology, Thyroid and Dental Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), India
| | - Ajay Kumar Singh
- Radiation Biosciences Division, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Delhi, India
| | - Velu Nair
- Army Hospital Research & Referral (AHRR), Delhi, India
| |
Collapse
|