1
|
Kut C, Quon H, Chen XS. Emerging Radiotherapy Technologies for Head and Neck Squamous Cell Carcinoma: Challenges and Opportunities in the Era of Immunotherapy. Cancers (Basel) 2024; 16:4150. [PMID: 39766050 PMCID: PMC11674243 DOI: 10.3390/cancers16244150] [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: 10/20/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Radiotherapy (RT) is an integral component in the multidisciplinary management of patients with head and neck squamous cell carcinoma (HNSCC). Significant advances have been made toward optimizing tumor control and toxicity profiles of RT for HNSCC in the past two decades. The development of intensity modulated radiotherapy (IMRT) and concurrent chemotherapy established the standard of care for most patients with locally advanced HNSCC around the turn of the century. More recently, selective dose escalation to the most radioresistant part of tumor and avoidance of the most critical substructures of organs at risk, often guided by functional imaging, allowed even further improvement in the therapeutic ratio of IMRT. Other highly conformal RT modalities, including intensity modulated proton therapy (IMPT) and stereotactic body radiotherapy (SBRT) are being increasingly utilized, although there are gaps in our understanding of the normal tissue complication probabilities and their relative biological effectiveness. There is renewed interest in spatially fractionated radiotherapy (SFRT), such as GRID and LATTICE radiotherapy, in both palliative and definitive settings. The emergence of immune checkpoint inhibitors (ICIs) has revolutionized the treatment of patients with recurrent and metastatic HNSCC. Novel RT modalities, including IMPT, SBRT, and SFRT, have the potential to reduce lymphopenia and immune suppression, stimulate anti-tumor immunity, and synergize with ICIs. The next frontier in the treatment of HNSCC may lie in the exploration of combined modality treatment with new RT technologies and ICIs.
Collapse
Affiliation(s)
- Carmen Kut
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21287, USA; (C.K.); (H.Q.)
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD 21287, USA; (C.K.); (H.Q.)
| | - Xuguang Scott Chen
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Coppes RP, van Dijk LV. Future of Team-based Basic and Translational Science in Radiation Oncology. Semin Radiat Oncol 2024; 34:370-378. [PMID: 39271272 DOI: 10.1016/j.semradonc.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
To further optimize radiotherapy, a more personalized treatment towards individual patient's risk profiles, dissecting both patient-specific tumor and normal tissue response to multimodality treatments is needed. Novel developments in radiobiology, using in vitro patient-specific complex tissue resembling 3D models and multiomics approaches at a spatial single-cell level, may provide unprecedented insight into the radiation responses of tumors and normal tissue. Here, we describe the necessary team effort, including all disciplines in radiation oncology, to integrate such data into clinical prediction models and link the relatively "big data" from the clinical practice, allowing accurate patient stratification for personalized treatment approaches.
Collapse
Affiliation(s)
- R P Coppes
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.; Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands..
| | - L V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
3
|
Camarda AM, Vincini MG, Russo S, Comi S, Emiro F, Bazani A, Ingargiola R, Vischioni B, Vecchi C, Volpe S, Orecchia R, Jereczek-Fossa BA, Orlandi E, Alterio D. Dosimetric and NTCP analyses for selecting parotid gland cancer patients for proton therapy. TUMORI JOURNAL 2024; 110:273-283. [PMID: 38769916 PMCID: PMC11295422 DOI: 10.1177/03008916241252544] [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: 08/17/2023] [Revised: 03/25/2024] [Accepted: 04/17/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE/OBJECTIVE To perform a dosimetric and a normal tissue complication probability (NTCP) comparison between intensity modulated proton therapy and photon volumetric modulated arc therapy in a cohort of patients with parotid gland cancers in a post-operative or radical setting. MATERIALS AND METHODS From May 2011 to September 2021, 37 parotid gland cancers patients treated at two institutions were eligible. Inclusion criteria were as follows: patients aged ⩾ 18 years, diagnosis of parotid gland cancers candidate for postoperative radiotherapy or definitive radiotherapy, presence of written informed consent for the use of anonymous data for research purposes. Organs at risk (OARs) were retrospectively contoured. Target coverage goal was defined as D95 > 98%. Six NTCP models were selected. NTCP profiles were calculated for each patient using an internally-developed Python script in RayStation TPS. Average differences in NTCP between photon and proton plans were tested for significance with a two-sided Wilcoxon signed-rank test. RESULTS Seventy-four plans were generated. A lower Dmean to the majority of organs at risk (inner ear, cochlea, oral cavity, pharyngeal constrictor muscles, contralateral parotid and submandibular gland) was obtained with intensity modulated proton therapy vs volumetric modulated arc therapy with statistical significance (p < .05). Ten (27%) patients had a difference in NTCP (photon vs proton plans) greater than 10% for hearing loss and tinnitus: among them, seven qualified for both endpoints, two patients for hearing loss only, and one for tinnitus. CONCLUSIONS In the current study, nearly one-third of patients resulted eligible for proton therapy and they were the most likely to benefit in terms of prevention of hearing loss and tinnitus.
Collapse
Affiliation(s)
- Anna Maria Camarda
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Maria Giulia Vincini
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Stefania Russo
- Medical Physics Unit, Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Stefania Comi
- Unit of Medical Physics, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesca Emiro
- Unit of Medical Physics, European Institute of Oncology IRCCS, Milan, Italy
| | - Alessia Bazani
- Medical Physics Unit, Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Rossana Ingargiola
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Barbara Vischioni
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
| | | | - Stefania Volpe
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Roberto Orecchia
- Scientific Directorate, European Institute of Oncology IRCCS, Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences,University of Pavia, Italy
| | - Daniela Alterio
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
4
|
Pillai S, Munguia-Lopez JG, Tran SD. Bioengineered Salivary Gland Microtissues─A Review of 3D Cellular Models and their Applications. ACS APPLIED BIO MATERIALS 2024; 7:2620-2636. [PMID: 38591955 DOI: 10.1021/acsabm.4c00028] [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: 04/10/2024]
Abstract
Salivary glands (SGs) play a vital role in maintaining oral health through the production and release of saliva. Injury to SGs can lead to gland hypofunction and a decrease in saliva secretion manifesting as xerostomia. While symptomatic treatments for xerostomia exist, effective permanent solutions are still lacking, emphasizing the need for innovative approaches. Significant progress has been made in the field of three-dimensional (3D) SG bioengineering for applications in gland regeneration. This has been achieved through a major focus on cell culture techniques, including soluble cues and biomaterial components of the 3D niche. Cells derived from both adult and embryonic SGs have highlighted key in vitro characteristics of SG 3D models. While still in its first decade of exploration, SG spheroids and organoids have so far served as crucial tools to study SG pathophysiology. This review, based on a literature search over the past decade, covers the importance of SG cell types in the realm of their isolation, sourcing, and culture conditions that modulate the 3D microenvironment. We discuss different biomaterials employed for SG culture and the current advances made in bioengineering SG models using them. The success of these 3D cellular models are further evaluated in the context of their applications in organ transplantation and in vitro disease modeling.
Collapse
Affiliation(s)
- Sangeeth Pillai
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| | - Jose G Munguia-Lopez
- Department of Mining and Materials Engineering, McGill University, 3610 University Street, Montreal, QC H3A 0C5, Canada
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
5
|
Hu C, Liao Z, Zhang L, Ma Z, Xiao C, Shao S, Gao Y. Alleviation of Splenic Injury by CB001 after Low-Dose Irradiation Mediated by NLRP3/Caspase-1-BAX/Caspase-3 Axis. Radiat Res 2024; 201:126-139. [PMID: 38154483 DOI: 10.1667/rade-22-00053.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 12/12/2023] [Indexed: 12/30/2023]
Abstract
Low-dose radiation has been extensively employed in clinical practice, including tumor immunotherapy, chronic inflammation treatment and nidus screening. However, the damage on the spleen caused by low-dose radiation significantly increases the risk of late infection-related mortality, and there is currently no corresponding protective strategy. In the present study, a novel compound preparation named CB001 mainly constituted of Acanthopanax senticosus (AS) and Oldenlandia diffusa (OD) was developed to alleviate splenic injury caused by fractionated low-dose exposures. As our results show that, white pulp atrophy and the excessive apoptosis in spleen tissue induced by radiation exposure were significantly ameliorated by CB001. Mechanistically, BAX-caspase-3 signaling and nucleotide-binding domain and leucine-rich-repeat-containing family pyrin 3 (NLRP3) inflammasome signaling were demonstrated to be involved in the radio-protective activity of CB001 with the selective activators. Furthermore, the crosstalk between apoptosis signaling and NLRP3 inflammasome signaling in mediating the radio-protective activity of CB001 was clarified, in which the pro-apoptotic protein BAX but not the anti-apoptotic protein Bcl2 was found to be downstream of NLRP3. Our study demonstrated that the use of a novel drug product CB001 can potentially facilitate the alleviation of radiation-induced splenic injury for patients receiving medical imaging diagnosis or fractionated radiation therapy.
Collapse
Affiliation(s)
- Changkun Hu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Zebin Liao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Liangliang Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Zengchun Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Chengrong Xiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| | - Shuai Shao
- National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Yue Gao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, P.R. China
| |
Collapse
|
6
|
Peters DR, Asher A, Conti A, Schiappacasse L, Daniel RT, Levivier M, Tuleasca C. Single fraction and hypofractionated radiosurgery for perioptic meningiomas-tumor control and visual outcomes: a systematic review and meta-analysis. Neurosurg Rev 2023; 46:287. [PMID: 37897519 DOI: 10.1007/s10143-023-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/21/2023] [Accepted: 10/22/2023] [Indexed: 10/30/2023]
Abstract
Perioptic meningiomas, defined as those that are less than 3 mm from the optic apparatus, are challenging to treat with stereotactic radiosurgery (SRS). Tumor control must be weighed against the risk of radiation-induced optic neuropathy (RION), as both tumor progression and RION can lead to visual decline. We performed a systematic review and meta-analysis of single fraction SRS and hypofractionated radiosurgery (hfRS) for perioptic meningiomas, evaluating tumor control and visual preservation rates. Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we reviewed articles published between 1968 and December 8, 2022. We retained 5 studies reporting 865 patients, 438 cases treated in single fraction, while 427 with hfRS. For single fraction SRS, the overall rate of tumor control was 95.1%, with actuarial rates at 5 and 10 years of 96% and 89%, respectively; tumor progression was 7.7%. The rate of visual stability was 90.4%, including visual improvement in 29.3%. The rate of visual decline was 9.6%, including blindness in 1.2%. For hfRS, the overall rate of tumor control was 95.6% (range 92.1-99.1, p < 0.001); tumor progression was 4.4% (range 0.9-7.9, p = 0.01). Overall rate of visual stability was 94.9% (range 90.9-98.9, p < 0.001), including visual improvement in 22.7% (range 5.0-40.3, p = 0.01); visual decline was 5.1% (range 1.1-9.1, p = 0.013). SRS is an effective and safe treatment option for perioptic meningiomas. Both hypofractionated regimens and single fraction SRS can be considered.
Collapse
Affiliation(s)
- David R Peters
- Carolina Neurosurgery & Spine Associates, Charlotte, NC, USA.
- Department of Neurosurgery, Atrium Health, Charlotte, NC, USA.
- Neurosurgery Service and Gamma Knife Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| | - Anthony Asher
- Carolina Neurosurgery & Spine Associates, Charlotte, NC, USA
- Department of Neurosurgery, Atrium Health, Charlotte, NC, USA
| | - Alfredo Conti
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Unit of Neurosurgery, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, Bologna, Italy
| | - Luis Schiappacasse
- Radiation Oncology Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Roy T Daniel
- Neurosurgery Service and Gamma Knife Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine (FBM), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Marc Levivier
- Neurosurgery Service and Gamma Knife Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine (FBM), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Constantin Tuleasca
- Neurosurgery Service and Gamma Knife Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine (FBM), University of Lausanne (UNIL), Lausanne, Switzerland
- Ecole Polytechnique Fédérale de Lausanne (EPFL, LTS-5), Lausanne, Switzerland
| |
Collapse
|
7
|
Zhang C, Xiang B. The underlying mechanisms and strategies of DNA damage and repair in radiation sialadenitis. Oral Dis 2023; 29:990-995. [PMID: 34773326 DOI: 10.1111/odi.14078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/18/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022]
Abstract
Radiation therapy is a critical strategy for the treatment of malignant tumors. X-ray external radiation has been successfully used to treat head and neck cancer. On the other hand, 131 I internal radiation has been effective in managing differentiated thyroid cancer. However, these therapies cause radiation damage to salivary glands. Radiation sialadenitis is the most common complication associated with radiotherapy applied to the head and neck and it severely affects patients' quality of life. Since DNA is the main intracellular target of radiation, and the integrity of the DNA structure is critical to genomic stability and the cellular survival of salivary glands, regulating radiation-induced DNA damage offers great promise in preventing and managing radiation sialadenitis. In this review, we summarize recent progress in DNA damage and repair in irradiated salivary glands.
Collapse
Affiliation(s)
- Chong Zhang
- Laboratory of Oral and Maxillofacial Disease, The Second Hospital of Dalian Medical University, Dalian, China
| | - Bin Xiang
- Laboratory of Oral and Maxillofacial Disease, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Liu Y, Liu Z, Hu L, He L, Yang L, Qin Z, Xie Y, Peng X, Dai L. Function of stem cells in radiation-induced damage. Int J Radiat Biol 2023; 99:1483-1494. [PMID: 36912588 DOI: 10.1080/09553002.2023.2188935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE The aim of this review is to discuss previous studies on the function of stem cells in radiation-induced damage, and the factors affecting these processes, in the hope of improving our understanding of the different stem cells and the communication networks surrounding them. This is essential for the development of effective stem cell-based therapies to regenerate or replace normal tissues damaged by radiation. CONCLUSION In salivary glands, senescence-associated cytokines and inflammation-associated cells have a greater effect on stem cells. In the intestinal glands, Paneth cells strongly affect stem cell-mediated tissue regeneration after radiation treatment. In the pancreas, β-cells as well as protein C receptor positive (Procr) cells are expected to be key cells in the treatment of diabetes. In the bone marrow, a variety of cytokines such as CXC-chemokine ligand 12 (CXCL12) and stem cell factor (SCF), contribute to the functional recovery of hematopoietic stem cells after irradiation.
Collapse
Affiliation(s)
- Yingtong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Hu
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lianlian Yang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuping Xie
- Department of Oncology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Sampadi B, Vermeulen S, Mišovic B, Boei JJ, Batth TS, Chang JG, Paulsen MT, Magnuson B, Schimmel J, Kool H, Olie CS, Everts B, Vertegaal ACO, Olsen JV, Ljungman M, Jeggo PA, Mullenders LHF, Vrieling H. Divergent Molecular and Cellular Responses to Low and High-Dose Ionizing Radiation. Cells 2022; 11:cells11233794. [PMID: 36497055 PMCID: PMC9739411 DOI: 10.3390/cells11233794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer risk after ionizing radiation (IR) is assumed to be linear with the dose; however, for low doses, definite evidence is lacking. Here, using temporal multi-omic systems analyses after a low (LD; 0.1 Gy) or a high (HD; 1 Gy) dose of X-rays, we show that, although the DNA damage response (DDR) displayed dose proportionality, many other molecular and cellular responses did not. Phosphoproteomics uncovered a novel mode of phospho-signaling via S12-PPP1R7, and large-scale dephosphorylation events that regulate mitotic exit control in undamaged cells and the G2/M checkpoint upon IR in a dose-dependent manner. The phosphoproteomics of irradiated DNA double-strand breaks (DSBs) repair-deficient cells unveiled extended phospho-signaling duration in either a dose-dependent (DDR signaling) or independent (mTOR-ERK-MAPK signaling) manner without affecting signal magnitude. Nascent transcriptomics revealed the transcriptional activation of genes involved in NRF2-regulated antioxidant defense, redox-sensitive ERK-MAPK signaling, glycolysis and mitochondrial function after LD, suggesting a prominent role for reactive oxygen species (ROS) in molecular and cellular responses to LD exposure, whereas DDR genes were prominently activated after HD. However, how and to what extent the observed dose-dependent differences in molecular and cellular responses may impact cancer development remain unclear, as the induction of chromosomal damage was found to be dose-proportional (10-200 mGy).
Collapse
Affiliation(s)
- Bharath Sampadi
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
- Correspondence: (B.S.); (H.V.)
| | - Sylvia Vermeulen
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Branislav Mišovic
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Jan J. Boei
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Tanveer S. Batth
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Jer-Gung Chang
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Michelle T. Paulsen
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brian Magnuson
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joost Schimmel
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Hanneke Kool
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Cyriel S. Olie
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
| | - Alfred C. O. Vertegaal
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Jesper V. Olsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Mats Ljungman
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Penny A. Jeggo
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN1 9RQ, UK
| | - Leon H. F. Mullenders
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya 464-8601, Japan
| | - Harry Vrieling
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
- Correspondence: (B.S.); (H.V.)
| |
Collapse
|
10
|
Serrano Martinez P, Maimets M, Bron R, van Os R, de Haan G, Pringle S, Coppes RP. Role of quiescent cells in the homeostatic maintenance of the adult submandibular salivary gland. iScience 2022; 25:105047. [PMID: 36147959 PMCID: PMC9485076 DOI: 10.1016/j.isci.2022.105047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/08/2022] [Accepted: 08/26/2022] [Indexed: 11/26/2022] Open
Abstract
Stem/progenitor cells are required for maintenance of salivary gland (SG) function and serve as untapped reservoirs to create functional cells. Despite recent advancements in the identification of stem/progenitor pools, in the submandibular gland (SMG), a knowledge gap remains. Furthermore, the contribution to adult SMG homeostasis of stem/progenitor cells originating from embryonic development is unclear. Here, we employ an H2B-GFP embryonic and adult pulse-and-chase system to characterize potential SMG stem/progenitor cells (SGSCs) based on quiescence at different stages. Phenotypical profiling of quiescent cells in the SMG revealed that label-retaining cells (LRCs) of embryonic or adult origin co-localized with CK8+ ductal or vimentin + mesenchymal, but not with CK5+ or CK14 + stem/progenitor cells. These SMG LRCs failed to self-renew in vitro while non-label retaining cells displayed differentiation and long-term expansion potential as organoids. Collectively, our data suggest that an active cycling population of cells is responsible for SMG homeostasis with organoid forming potential. Embryonic quiescent cells do not retain stemness in the adult submandibular gland (SMG) Postnatal quiescent cells do not exhibit stem/progenitor cell potency in the adult SMG Quiescent cells do not contribute to the homeostatic maintenance of the murine SMG Adult murine SMG stem/progenitor cells are likely to be an actively cycling population
Collapse
Affiliation(s)
- Paola Serrano Martinez
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB Groningen, the Netherlands
| | - Martti Maimets
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB Groningen, the Netherlands
| | - Reinier Bron
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB Groningen, the Netherlands.,Department of Biomedical Engineering, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Ronald van Os
- Department of Biology of Aging, Section Stem Cell Biology, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Gerald de Haan
- Department of Biology of Aging, Section Stem Cell Biology, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Robert P Coppes
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
11
|
Blake SW. Can dose convolution modelling explain bath and shower effects in rat spinal cord? Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac5c8e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/10/2022] [Indexed: 12/24/2022]
Abstract
Abstract
Objective. ‘Bath and shower’ effects were first seen in proton irradiations of rat spinal cord, where a low dose ‘bath’ reduced the smaller field ‘shower’ dose needed for limb paralysis giving the appearance of sensitisation of the cord or disproportionate response. This was difficult to reconcile with existing tissue complication models. The purpose of this investigation is to explore a different approach using a dose convolution algorithm to model the 50% isoeffect endpoint. Approach. Bath and shower dose distributions were convolved with Gaussian functions with widths specified by the σ parameter. The hypothesis was that the maximum value from the convolved distributions was constant for isoeffect across the modelled scenarios. A simpler field length dependent relative biological effectiveness (FLRBE) approach was also used for a subset of the data which gave results independent of σ. Main results. The maximum values from the convolved distributions were constant within ±17% across the bath and shower experiments for σ = 3.5 mm, whereas the maximum dose varied by a factor of four. The FLRBE results were also within ±14% confirming the validity of the dose convolution approach. Significance. A simple approach using dose convolution modelling of the 50% isotoxicity gave compelling consistency with the full range of bath and shower results, while the FLRBE approach confirmed the results for the symmetric field data. Convolution modelling and the effect of time interval were consistent with a signalling factor diffusion mechanism such as the ‘bystander effect’. The results suggest biological effectiveness is reduced for very small field sizes, requiring a higher isoeffect dose. By implication, the bath dose does not sensitise the cord to the shower dose; when biological effectiveness is accounted for, a small increase in the bath dose requires a significantly larger reduction in shower dose for isoeffect.
Collapse
|
12
|
Unlaid Eggs: Ovarian Damage after Low-Dose Radiation. Cells 2022; 11:cells11071219. [PMID: 35406783 PMCID: PMC8997758 DOI: 10.3390/cells11071219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/24/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
The total body irradiation of lymphomas and co-irradiation in the treatment of adjacent solid tumors can lead to a reduced ovarian function, premature ovarian insufficiency, and menopause. A small number of studies has assessed the radiation-induced damage of primordial follicles in animal models and humans. Studies are emerging that evaluate radiation-induced damage to the surrounding ovarian tissue including stromal and immune cells. We reviewed basic laboratory work to assess the current state of knowledge and to establish an experimental setting for further studies in animals and humans. The experimental approaches were mostly performed using mouse models. Most studies relied on single doses as high as 1 Gy, which is considered to cause severe damage to the ovary. Changes in the ovarian reserve were related to the primordial follicle count, providing reproducible evidence that radiation with 1 Gy leads to a significant depletion. Radiation with 0.1 Gy mostly did not show an effect on the primordial follicles. Fewer data exist on the effects of radiation on the ovarian microenvironment including theca-interstitial, immune, endothelial, and smooth muscle cells. We concluded that a mouse model would provide the most reliable model to study the effects of low-dose radiation. Furthermore, both immunohistochemistry and fluorescence-activated cell sorting (FACS) analyses were valuable to analyze not only the germ cells but also the ovarian microenvironment.
Collapse
|
13
|
Steenbakkers RJHM, van Rijn-Dekker MI, Stokman MA, Kierkels RGJ, van der Schaaf A, van den Hoek JGM, Bijl HP, Kramer MCA, Coppes RP, Langendijk JA, van Luijk P. Parotid Gland Stem Cell Sparing Radiation Therapy for Patients With Head and Neck Cancer: A Double-Blind Randomized Controlled Trial. Int J Radiat Oncol Biol Phys 2022; 112:306-316. [PMID: 34563635 DOI: 10.1016/j.ijrobp.2021.09.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE Radiation therapy for head and neck cancer frequently leads to salivary gland damage and subsequent xerostomia. The radiation response of the parotid glands of rats, mice, and patients critically depends on dose to parotid gland stem cells, mainly located in the gland's main ducts (stem cell rich [SCR] region). Therefore, this double-blind randomized controlled trial aimed to test the hypothesis that parotid gland stem cell sparing radiation therapy preserves parotid gland function better than currently used whole parotid gland sparing radiation therapy. METHODS AND MATERIALS Patients with head and neck cancer (n = 102) treated with definitive radiation therapy were randomized between standard parotid-sparing and stem cell sparing (SCS) techniques. The primary endpoint was >75% reduction in parotid gland saliva production compared with pretreatment production (FLOW12M). Secondary endpoints were several aspects of xerostomia 12 months after treatment. RESULTS Fifty-four patients were assigned to the standard arm and 48 to the SCS arm. Only dose to the SCR regions (contralateral 16 and 11 Gy [P = .004] and ipsilateral 26 and 16 Gy [P = .001] in the standard and SCS arm, respectively) and pretreatment patient-rated daytime xerostomia (35% and 13% [P = .01] in the standard and SCS arm, respectively) differed significantly between the arms. In the SCS arm, 1 patient (2.8%) experienced FLOW12M compared with 2 (4.9%) in the standard arm (P = 1.00). However, a trend toward better relative parotid gland salivary function in favor of SCS radiation therapy was shown. Moreover, multivariable analysis showed that mean contralateral SCR region dose was the strongest dosimetric predictor for moderate-to-severe patient-rated daytime xerostomia and grade ≥2 physician-rated xerostomia, the latter including reported alteration in diet. CONCLUSIONS No significantly better parotid function was observed in SCS radiation therapy. However, additional multivariable analysis showed that dose to the SCR region was more predictive of the development of parotid gland function-related xerostomia endpoints than dose to the entire parotid gland.
Collapse
Affiliation(s)
- Roel J H M Steenbakkers
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Maria I van Rijn-Dekker
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Monique A Stokman
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Roel G J Kierkels
- Department of Radiation Oncology, Radiotherapiegroep, Deventer, The Netherlands
| | - Arjen van der Schaaf
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johanna G M van den Hoek
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hendrik P Bijl
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maria C A Kramer
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robert P Coppes
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Biomedical Sciences of Cell and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - 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.
| |
Collapse
|
14
|
D'Auria Vieira de Godoy PR, Nakamura A, Khavari AP, Sangsuwan T, Haghdoost S. Effect of dose and dose rate of gamma irradiation on the formation of micronuclei in bone marrow cells isolated from whole-body-irradiated mice. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2021; 62:422-427. [PMID: 34296472 DOI: 10.1002/em.22453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/10/2021] [Accepted: 07/18/2021] [Indexed: 06/13/2023]
Abstract
It is well-known that the cytotoxicity and mutagenic effects of high dose rate (HDR) ionizing radiation (IR) are increased by increasing the dose but less is known about the effects of chronic low dose rate (LDR). In vitro, we have shown that in addition to the immediate interaction of IR with DNA (the direct and indirect effects), low doses and chronic LDR exposure induce endogenous oxidative stress. During elevated oxidative stress, reactive oxygen species (ROS) react with DNA modifying its structure. Here, BL6 mice were exposed to IR at LDR and HDR and were then sacrificed 3 hours and 3 weeks after exposure to examine early and late effects of IR. The levels of micronuclei, MN, were determined in bone marrow cells. Our data indicate that the effects of 200 mGy on MN-induction are transient, but 500 and 1000 mGy (both HDR and LDR) lead to increased levels of MN up to 3 weeks after the exposure.
Collapse
Affiliation(s)
| | - Ayumi Nakamura
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Ali Pour Khavari
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Traimate Sangsuwan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Siamak Haghdoost
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- University of Caen Normandy, ARIA-CIMAP Laboratory, Campus Jules Horowitz, Caen, France
| |
Collapse
|
15
|
Hanania AN, Zhang X, Gunn GB, Rosenthal DI, Garden AS, Fuller CD, Phan J, Reddy JP, Moreno A, Chronowski G, Shah S, Ausat N, Hanna E, Ferrarotto R, Frank SJ. Proton Therapy for Major Salivary Gland Cancer: Clinical Outcomes. Int J Part Ther 2021; 8:261-272. [PMID: 34285952 PMCID: PMC8270094 DOI: 10.14338/ijpt-20-00044.1] [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] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 11/21/2022] Open
Abstract
Purpose To report clinical outcomes in terms of disease control and toxicity in patients with major salivary gland cancers (SGCs) treated with proton beam therapy. Materials and Methods Clinical and dosimetric characteristics of patients with SGCs treated from August 2011 to February 2020 on an observational, prospective, single-institution protocol were abstracted. Local control and overall survival were calculated by the Kaplan-Meier method. During radiation, weekly assessments of toxicity were obtained, and for patients with ≥ 90 days of follow-up, late toxicity was assessed. Results Seventy-two patients were identified. Median age was 54 years (range, 23-87 years). Sixty-three patients (88%) received postoperative therapy, and nine patients (12%) were treated definitively. Twenty-six patients (36%) received concurrent chemotherapy. Nine patients (12%) had received prior radiation. All (99%) but one patient received unilateral treatment with a median dose of 64 GyRBE (relative biological effectiveness) (interquartile range [IQR], 60-66), and 53 patients (74%) received intensity-modulated proton therapy with either single-field or multifield optimization. The median follow-up time was 30 months. Two-year local control and overall survival rates were 96% (95% confidence interval [CI] 85%-99%) and 89% (95% CI 76%-95%], respectively. Radiation dermatitis was the predominant grade-3 toxicity (seen in 21% [n = 15] of the patients), and grade ≥ 2 mucositis was rare (14%; n = 10 patients). No late-grade ≥ 3 toxicities were reported. Conclusion Proton beam therapy for treatment of major SGCs manifests in low rates of acute mucosal toxicity. In addition, the current data suggest a high rate of local control and minimal late toxicity.
Collapse
Affiliation(s)
- Alexander N Hanania
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaodong Zhang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David I Rosenthal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam S Garden
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C David Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack Phan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jay P Reddy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy Moreno
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Chronowski
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shalin Shah
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Noveen Ausat
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ehab Hanna
- Department of Head and Neck Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renata Ferrarotto
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven J Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
16
|
Bunevicius A, Anand RK, Suleiman M, Nabeel AM, Reda WA, Tawadros SR, Abdelkarim K, El-Shehaby AMN, Emad RM, Chytka T, Liscak R, Sheehan K, Sheehan D, Caceres MP, Mathieu D, Lee CC, Yang HC, Picozzi P, Franzini A, Attuati L, Speckter H, Olivo J, Patel S, Cifarelli CP, Cifarelli DT, Hack JD, Strickland BA, Zada G, Chang EL, Fakhoury KR, Rusthoven CG, Warnick RE, Sheehan J. Stereotactic Radiosurgery for Perioptic Meningiomas: An International, Multicenter Study. Neurosurgery 2021; 88:828-837. [PMID: 33475718 DOI: 10.1093/neuros/nyaa544] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/10/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Stereotactic radiosurgery (SRS) is increasingly used for management of perioptic meningiomas. OBJECTIVE To study the safety and effectiveness of SRS for perioptic meningiomas. METHODS From 12 institutions participating in the International Radiosurgery Research Foundation (IRRF), we retrospectively assessed treatment parameters and outcomes following SRS for meningiomas located within 3 mm of the optic apparatus. RESULTS A total of 438 patients (median age 51 yr) underwent SRS for histologically confirmed (29%) or radiologically suspected (71%) perioptic meningiomas. Median treatment volume was 8.01 cm3. Median prescription dose was 12 Gy, and median dose to the optic apparatus was 8.50 Gy. A total of 405 patients (93%) underwent single-fraction SRS and 33 patients (7%) underwent hypofractionated SRS. During median imaging follow-up of 55.6 mo (range: 3.15-239 mo), 33 (8%) patients experienced tumor progression. Actuarial 5-yr and 10-yr progression-free survival was 96% and 89%, respectively. Prescription dose of ≥12 Gy (HR: 0.310; 95% CI [0.141-0.679], P = .003) and single-fraction SRS (HR: 0.078; 95% CI [0.016-0.395], P = .002) were associated with improved tumor control. A total of 31 (10%) patients experienced visual decline, with actuarial 5-yr and 10-yr post-SRS visual decline rates of 9% and 21%, respectively. Maximum dose to the optic apparatus ≥10 Gy (HR = 2.370; 95% CI [1.086-5.172], P = .03) and tumor progression (HR = 4.340; 95% CI [2.070-9.097], P < .001) were independent predictors of post-SRS visual decline. CONCLUSION SRS provides durable tumor control and quite acceptable rates of vision preservation in perioptic meningiomas. Margin dose of ≥12 Gy is associated with improved tumor control, while a dose to the optic apparatus of ≥10 Gy and tumor progression are associated with post-SRS visual decline.
Collapse
Affiliation(s)
- Adomas Bunevicius
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | | | - Mohanad Suleiman
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | - Ahmed M Nabeel
- Gamma Knife Center Cairo, Nasser Institute Hospital, Cairo, Egypt.,Neurosurgery Department, Benha University, Qalubya, Egypt
| | - Wael A Reda
- Gamma Knife Center Cairo, Nasser Institute Hospital, Cairo, Egypt.,Neurosurgery Department, Ain Shams University, Cairo, Egypt
| | - Sameh R Tawadros
- Gamma Knife Center Cairo, Nasser Institute Hospital, Cairo, Egypt.,Neurosurgery Department, Ain Shams University, Cairo, Egypt
| | - Khaled Abdelkarim
- Gamma Knife Center Cairo, Nasser Institute Hospital, Cairo, Egypt.,Clinical Oncology Department, Ain Shams University, Cairo, Egypt
| | - Amr M N El-Shehaby
- Gamma Knife Center Cairo, Nasser Institute Hospital, Cairo, Egypt.,Neurosurgery Department, Ain Shams University, Cairo, Egypt
| | - Reem M Emad
- Gamma Knife Center Cairo, Nasser Institute Hospital, Cairo, Egypt.,Radiation Oncology Department, National Cancer Institute, Cairo University, Giza, Egypt
| | - Tomas Chytka
- Stereotactic and Radiation Neurosurgery Department, Na Homolce Hospital, Prague, Czech Republic
| | - Roman Liscak
- Stereotactic and Radiation Neurosurgery Department, Na Homolce Hospital, Prague, Czech Republic
| | - Kimball Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | - Darrah Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | - Marco Perez Caceres
- Department of Neurosurgery, Université de Sherbrooke, Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - David Mathieu
- Department of Neurosurgery, Université de Sherbrooke, Centre de recherche du CHUS, Sherbrooke, Québec, Canada
| | - Cheng-Chia Lee
- Department of Neurosurgery, Neurological Institute, Taipei Veteran General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Huai-Che Yang
- Department of Neurosurgery, Neurological Institute, Taipei Veteran General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Piero Picozzi
- Department of Neurosurgery, Humanitas Clinical and Research Center - IRCCS, Milan, Italy
| | - Andrea Franzini
- Department of Neurosurgery, Humanitas Clinical and Research Center - IRCCS, Milan, Italy
| | - Luca Attuati
- Department of Neurosurgery, Humanitas Clinical and Research Center - IRCCS, Milan, Italy
| | - Herwin Speckter
- Centro Gamma Knife Dominicano and CEDIMAT Radiology Department, Santo Domingo, Dominican Republic
| | - Jeremy Olivo
- Centro Gamma Knife Dominicano and CEDIMAT Radiology Department, Santo Domingo, Dominican Republic
| | - Samir Patel
- Division of Radiation Oncology, Department of Oncology, University of Alberta, Edmonton, Canada
| | - Christopher P Cifarelli
- Department of Neurosurgery, West Virginia University, Morgantown, West Virginia.,Department of Radiation Oncology, West Virginia University, Morgantown, West Virginia
| | - Daniel T Cifarelli
- Department of Neurosurgery, West Virginia University, Morgantown, West Virginia
| | - Joshua D Hack
- Department of Radiation Oncology, West Virginia University, Morgantown, West Virginia
| | - Ben A Strickland
- Department of Neurosurgery, University of Southern California, Los Angeles, California
| | - Gabriel Zada
- Department of Neurosurgery, University of Southern California, Los Angeles, California
| | - Eric L Chang
- Department of Radiation Oncology, University of Southern California, Los Angeles, California
| | - Kareem R Fakhoury
- Department of Radiation Oncology, University of Colorado, Denver, Colorado
| | - Chad G Rusthoven
- Department of Radiation Oncology, University of Colorado, Denver, Colorado
| | - Ronald E Warnick
- Gamma Knife Center, Jewish Hospital, Mayfield Clinic, Cincinnati, Ohio
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
17
|
Rae C, Amato F, Braconi C. Patient-Derived Organoids as a Model for Cancer Drug Discovery. Int J Mol Sci 2021; 22:ijms22073483. [PMID: 33801782 PMCID: PMC8038043 DOI: 10.3390/ijms22073483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
In the search for the ideal model of tumours, the use of three-dimensional in vitro models is advancing rapidly. These are intended to mimic the in vivo properties of the tumours which affect cancer development, progression and drug sensitivity, and take into account cell–cell interactions, adhesion and invasiveness. Importantly, it is hoped that successful recapitulation of the structure and function of the tissue will predict patient response, permitting the development of personalized therapy in a timely manner applicable to the clinic. Furthermore, the use of co-culture systems will allow the role of the tumour microenvironment and tissue–tissue interactions to be taken into account and should lead to more accurate predictions of tumour development and responses to drugs. In this review, the relative merits and limitations of patient-derived organoids will be discussed compared to other in vitro and ex vivo cancer models. We will focus on their use as models for drug testing and personalized therapy and how these may be improved. Developments in technology will also be considered, including the use of microfluidics, 3D bioprinting, cryopreservation and circulating tumour cell-derived organoids. These have the potential to enhance the consistency, accessibility and availability of these models.
Collapse
Affiliation(s)
- Colin Rae
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (C.R.); (F.A.)
| | - Francesco Amato
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (C.R.); (F.A.)
| | - Chiara Braconi
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (C.R.); (F.A.)
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, UK
- Correspondence:
| |
Collapse
|
18
|
Paganetti H, Beltran C, Both S, Dong L, Flanz J, Furutani K, Grassberger C, Grosshans DR, Knopf AC, Langendijk JA, Nystrom H, Parodi K, Raaymakers BW, Richter C, Sawakuchi GO, Schippers M, Shaitelman SF, Teo BKK, Unkelbach J, Wohlfahrt P, Lomax T. Roadmap: proton therapy physics and biology. Phys Med Biol 2021; 66. [DOI: 10.1088/1361-6560/abcd16] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
|
19
|
Voshart DC, Wiedemann J, van Luijk P, Barazzuol L. Regional Responses in Radiation-Induced Normal Tissue Damage. Cancers (Basel) 2021; 13:cancers13030367. [PMID: 33498403 PMCID: PMC7864176 DOI: 10.3390/cancers13030367] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Normal tissue side effects remain a major concern in radiotherapy. The improved precision of radiation dose delivery of recent technological developments in radiotherapy has the potential to reduce the radiation dose to organ regions that contribute the most to the development of side effects. This review discusses the contribution of regional variation in radiation responses in several organs. In the brain, various regions were found to contribute to radiation-induced neurocognitive dysfunction. In the parotid gland, the region containing the major ducts was found to be critical in hyposalivation. The heart and lung were each found to exhibit regional responses while also mutually affecting each other's response to radiation. Sub-structures critical for the development of side effects were identified in the pancreas and bladder. The presence of these regional responses is based on a non-uniform distribution of target cells or sub-structures critical for organ function. These characteristics are common to most organs in the body and we therefore hypothesize that regional responses in radiation-induced normal tissue damage may be a shared occurrence. Further investigations will offer new opportunities to reduce normal tissue side effects of radiotherapy using modern and high-precision technologies.
Collapse
Affiliation(s)
- Daniëlle C. Voshart
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Julia Wiedemann
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Peter van Luijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (P.v.L.); (L.B.)
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (P.v.L.); (L.B.)
| |
Collapse
|
20
|
Current and Future Perspectives of the Use of Organoids in Radiobiology. Cells 2020; 9:cells9122649. [PMID: 33317153 PMCID: PMC7764598 DOI: 10.3390/cells9122649] [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: 11/23/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The majority of cancer patients will be treated with radiotherapy, either alone or together with chemotherapy and/or surgery. Optimising the balance between tumour control and the probability of normal tissue side effects is the primary goal of radiation treatment. Therefore, it is imperative to understand the effects that irradiation will have on both normal and cancer tissue. The more classical lab models of immortal cell lines and in vivo animal models have been fundamental to radiobiological studies to date. However, each of these comes with their own limitations and new complementary models are required to fill the gaps left by these traditional models. In this review, we discuss how organoids, three-dimensional tissue-resembling structures derived from tissue-resident, embryonic or induced pluripotent stem cells, overcome the limitations of these models and thus have a growing importance in the field of radiation biology research. The roles of organoids in understanding radiation-induced tissue responses and in moving towards precision medicine are examined. Finally, the limitations of organoids in radiobiology and the steps being made to overcome these limitations are considered.
Collapse
|
21
|
Patera V, Prezado Y, Azaiez F, Battistoni G, Bettoni D, Brandenburg S, Bugay A, Cuttone G, Dauvergne D, de France G, Graeff C, Haberer T, Inaniwa T, Incerti S, Nasonova E, Navin A, Pullia M, Rossi S, Vandevoorde C, Durante M. Biomedical Research Programs at Present and Future High-Energy Particle Accelerators. FRONTIERS IN PHYSICS 2020; 8:00380. [PMID: 33224942 PMCID: PMC7116397 DOI: 10.3389/fphy.2020.00380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Biomedical applications at high-energy particle accelerators have always been an important section of the applied nuclear physics research. Several new facilities are now under constructions or undergoing major upgrades. While the main goal of these facilities is often basic research in nuclear physics, they acknowledge the importance of including biomedical research programs and of interacting with other medical accelerator facilities providing patient treatments. To harmonize the programs, avoid duplications, and foster collaboration and synergism, the International Biophysics Collaboration is providing a platform to several accelerator centers with interest in biomedical research. In this paper, we summarize the programs of various facilities in the running, upgrade, or construction phase.
Collapse
Affiliation(s)
- Vincenzo Patera
- Dipartimento di Scienze di Base e Applicate per l’Ingegneria, University “La Sapienza”, Rome, Italy
| | | | | | | | | | | | | | | | - Denis Dauvergne
- Université Grenoble-Alpes, CNRS/IN2P3, UMR5821, LPSC, GDR MI2B, LabEx PRIMES, Grenoble, France
| | | | - Christian Graeff
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | | | - Sebastien Incerti
- Université de Bordeaux, CNRS/IN2P3, UMR5797, Centre d’Études Nucléaires de Bordeaux Gradignan, Gradignan, France
| | | | | | | | | | | | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
- Institut für Festkörperphysik, Technische Universität Darmstadt, Darmstadt, Germany
- Correspondence: Marco Durante,
| |
Collapse
|
22
|
Cellular senescence contributes to radiation-induced hyposalivation by affecting the stem/progenitor cell niche. Cell Death Dis 2020; 11:854. [PMID: 33056980 PMCID: PMC7566836 DOI: 10.1038/s41419-020-03074-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023]
Abstract
Radiotherapy for head and neck cancer is associated with impairment of salivary gland function and consequent xerostomia, which has a devastating effect on the quality of life of the patients. The mechanism of radiation-induced salivary gland damage is not completely understood. Cellular senescence is a permanent state of cell cycle arrest accompanied by a secretory phenotype which contributes to inflammation and tissue deterioration. Genotoxic stresses, including radiation-induced DNA damage, are known to induce a senescence response. Here, we show that radiation induces cellular senescence preferentially in the salivary gland stem/progenitor cell niche of mouse models and patients. Similarly, salivary gland-derived organoids show increased expression of senescence markers and pro-inflammatory senescence-associated secretory phenotype (SASP) factors after radiation exposure. Clearance of senescent cells by selective removal of p16Ink4a-positive cells by the drug ganciclovir or the senolytic drug ABT263 lead to increased stem cell self-renewal capacity as measured by organoid formation efficiency. Additionally, pharmacological treatment with ABT263 in mice irradiated to the salivary glands mitigates tissue degeneration, thus preserving salivation. Our data suggest that senescence in the salivary gland stem/progenitor cell niche contributes to radiation-induced hyposalivation. Pharmacological targeting of senescent cells may represent a therapeutic strategy to prevent radiotherapy-induced xerostomia.
Collapse
|
23
|
Hacker BC, Rafat M. Organoids as Complex In Vitro Models for Studying Radiation-Induced Cell Recruitment. Cell Mol Bioeng 2020; 13:341-357. [PMID: 32952734 PMCID: PMC7479086 DOI: 10.1007/s12195-020-00625-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/10/2020] [Indexed: 01/01/2023] Open
Abstract
Patients with triple negative breast cancer (TNBC) typically receive chemotherapy, surgery, and radiation therapy. Although this treatment improves prognosis for most patients, some patients continue to experience recurrence within 5 years. Preclinical studies have shown that immune cell infiltration at the irradiated site may play a significant role in tumor cell recruitment; however, little is known about the mechanisms that govern this process. This lack of knowledge highlights the need to evaluate radiation-induced cell infiltration with models that have controllable variables and maintain biological integrity. Mammary organoids are multicellular three-dimensional (3D) in vitro models, and they have been used to examine many aspects of mammary development and tumorigenesis. Organoids are also emerging as a powerful tool to investigate normal tissue radiation damage. In this review, we evaluate recent advances in mammary organoid technology, consider the advantages of using organoids to study radiation response, and discuss future directions for the applications of this technique.
Collapse
Affiliation(s)
- Benjamin C. Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
24
|
Preclinical models of head and neck squamous cell carcinoma for a basic understanding of cancer biology and its translation into efficient therapies. CANCERS OF THE HEAD & NECK 2020; 5:9. [PMID: 32714605 PMCID: PMC7376675 DOI: 10.1186/s41199-020-00056-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
Comprehensive molecular characterization of head and neck squamous cell carcinoma (HNSCC) has led to the identification of distinct molecular subgroups with fundamental differences in biological properties and clinical behavior. Despite improvements in tumor classification and increased understanding about the signaling pathways involved in neoplastic transformation and disease progression, current standard-of-care treatment for HNSCC mostly remains to be based on a stage-dependent strategy whereby all patients at the same stage receive the same treatment. Preclinical models that closely resemble molecular HNSCC subgroups that can be exploited for dissecting the biological function of genetic variants and/or altered gene expression will be highly valuable for translating molecular findings into improved clinical care. In the present review, we merge and discuss existing and new information on established cell lines, primary two- and three-dimensional ex vivo tumor cultures from HNSCC patients, and animal models. We review their value in elucidating the basic biology of HNSCC, molecular mechanisms of treatment resistance and their potential for the development of novel molecularly stratified treatment.
Collapse
|
25
|
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: 110] [Impact Index Per Article: 22.0] [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
|
26
|
Barazzuol L, Coppes RP, van Luijk P. Prevention and treatment of radiotherapy-induced side effects. Mol Oncol 2020; 14:1538-1554. [PMID: 32521079 PMCID: PMC7332214 DOI: 10.1002/1878-0261.12750] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
Radiotherapy remains a mainstay of cancer treatment, being used in roughly 50% of patients. The precision with which the radiation dose can be delivered is rapidly improving. This precision allows the more accurate targeting of radiation dose to the tumor and reduces the amount of surrounding normal tissue exposed. Although this often reduces the unwanted side effects of radiotherapy, we still need to further improve patients' quality of life and to escalate radiation doses to tumors when necessary. High-precision radiotherapy forces one to choose which organ or functional organ substructures should be spared. To be able to make such choices, we urgently need to better understand the molecular and physiological mechanisms of normal tissue responses to radiotherapy. Currently, oversimplified approaches using constraints on mean doses, and irradiated volumes of normal tissues are used to plan treatments with minimized risk of radiation side effects. In this review, we discuss the responses of three different normal tissues to radiotherapy: the salivary glands, cardiopulmonary system, and brain. We show that although they may share very similar local cellular processes, they respond very differently through organ-specific, nonlocal mechanisms. We also discuss how a better knowledge of these mechanisms can be used to treat or to prevent the effects of radiotherapy on normal tissue and to optimize radiotherapy delivery.
Collapse
Affiliation(s)
- Lara Barazzuol
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Rob P. Coppes
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Peter van Luijk
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
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
|
Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther 2020; 5:60. [PMID: 32355263 PMCID: PMC7192953 DOI: 10.1038/s41392-020-0150-x] [Citation(s) in RCA: 566] [Impact Index Per Article: 113.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/20/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy is one of the most common countermeasures for treating a wide range of tumors. However, the radioresistance of cancer cells is still a major limitation for radiotherapy applications. Efforts are continuously ongoing to explore sensitizing targets and develop radiosensitizers for improving the outcomes of radiotherapy. DNA double-strand breaks are the most lethal lesions induced by ionizing radiation and can trigger a series of cellular DNA damage responses (DDRs), including those helping cells recover from radiation injuries, such as the activation of DNA damage sensing and early transduction pathways, cell cycle arrest, and DNA repair. Obviously, these protective DDRs confer tumor radioresistance. Targeting DDR signaling pathways has become an attractive strategy for overcoming tumor radioresistance, and some important advances and breakthroughs have already been achieved in recent years. On the basis of comprehensively reviewing the DDR signal pathways, we provide an update on the novel and promising druggable targets emerging from DDR pathways that can be exploited for radiosensitization. We further discuss recent advances identified from preclinical studies, current clinical trials, and clinical application of chemical inhibitors targeting key DDR proteins, including DNA-PKcs (DNA-dependent protein kinase, catalytic subunit), ATM/ATR (ataxia-telangiectasia mutated and Rad3-related), the MRN (MRE11-RAD50-NBS1) complex, the PARP (poly[ADP-ribose] polymerase) family, MDC1, Wee1, LIG4 (ligase IV), CDK1, BRCA1 (BRCA1 C terminal), CHK1, and HIF-1 (hypoxia-inducible factor-1). Challenges for ionizing radiation-induced signal transduction and targeted therapy are also discussed based on recent achievements in the biological field of radiotherapy.
Collapse
Affiliation(s)
- Rui-Xue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, 410078, Changsha, People's Republic of China
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, 100850, Beijing, People's Republic of China.
- Institute for Chemical Carcinogenesis, State Key Laboratory of Respiratory, Guangzhou Medical University, 511436, Guangzhou, People's Republic of China.
| |
Collapse
|
29
|
Dünker N, Jendrossek V. Implementation of the Chick Chorioallantoic Membrane (CAM) Model in Radiation Biology and Experimental Radiation Oncology Research. Cancers (Basel) 2019; 11:cancers11101499. [PMID: 31591362 PMCID: PMC6826367 DOI: 10.3390/cancers11101499] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) is part of standard cancer treatment. Innovations in treatment planning and increased precision in dose delivery have significantly improved the therapeutic gain of radiotherapy but are reaching their limits due to biologic constraints. Thus, a better understanding of the complex local and systemic responses to RT and of the biological mechanisms causing treatment success or failure is required if we aim to define novel targets for biological therapy optimization. Moreover, optimal treatment schedules and prognostic biomarkers have to be defined for assigning patients to the best treatment option. The complexity of the tumor environment and of the radiation response requires extensive in vivo experiments for the validation of such treatments. So far in vivo investigations have mostly been performed in time- and cost-intensive murine models. Here we propose the implementation of the chick chorioallantoic membrane (CAM) model as a fast, cost-efficient model for semi high-throughput preclinical in vivo screening of the modulation of the radiation effects by molecularly targeted drugs. This review provides a comprehensive overview on the application spectrum, advantages and limitations of the CAM assay and summarizes current knowledge of its applicability for cancer research with special focus on research in radiation biology and experimental radiation oncology.
Collapse
Affiliation(s)
- Nicole Dünker
- Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| |
Collapse
|
30
|
Nagle PW, van Goethem MJ, Kempers M, Kiewit H, Knopf A, Langendijk JA, Brandenburg S, van Luijk P, Coppes RP. In vitro biological response of cancer and normal tissue cells to proton irradiation not affected by an added magnetic field. Radiother Oncol 2019; 137:125-129. [PMID: 31085392 DOI: 10.1016/j.radonc.2019.04.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 01/26/2023]
Abstract
To optimize beam delivery and conformality of proton therapy, MRI integration has been proposed. Therefore, we investigated if proton irradiation in a magnetic field would change biological responses. Our data in cancer cell lines and stem cell-derived organoid models suggest that a magnetic field does not modify the biological response.
Collapse
Affiliation(s)
- Peter W Nagle
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Marc-Jan van Goethem
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands; KVI Center for Advanced Radiation Technology, University of Groningen, The Netherlands
| | - Marco Kempers
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Harry Kiewit
- KVI Center for Advanced Radiation Technology, University of Groningen, The Netherlands
| | - Antje Knopf
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Johannes A Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Sytze Brandenburg
- KVI Center for Advanced Radiation Technology, University of Groningen, The Netherlands
| | - Peter van Luijk
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Rob P Coppes
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands.
| |
Collapse
|
31
|
Peitzsch C, Kurth I, Ebert N, Dubrovska A, Baumann M. Cancer stem cells in radiation response: current views and future perspectives in radiation oncology. Int J Radiat Biol 2019; 95:900-911. [PMID: 30897014 DOI: 10.1080/09553002.2019.1589023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose: Despite technological improvement and advances in biology-driven patient stratification, many patients still fail radiotherapy resulting in loco-regional and distant recurrence. Tumor heterogeneity remains a key challenge to effective cancer treatment, and reliable stratification of cancer patients for prediction of outcomes is highly important. Intratumoral heterogeneity is manifested at the different levels, including different tumorigenic properties of cancer cells. Since John Dick et al. isolated leukemia initiating cells in 1990, the populations of tumor initiating or cancer stem cells (CSCs) were identified and characterized also for a broad spectrum of solid tumor types. The properties of CSCs are of considerable clinical relevance: CSCs have self-renewal and tumor initiating potential, and the metastases are initiated by the CSC clones with the ability to disseminate from the primary tumor site. Conclusion: Evidence from both, experimental and clinical studies demonstrates that the probability of achieving local tumor control by radiation therapy depends on the complete eradication of CSC populations. The number, properties and molecular signature of CSCs are highly predictive for clinical outcome of radiotherapy, whereas targeted therapies against CSCs combined with conventional treatment are expected to provide an improved clinical response and prevent tumor relapse. In this review, we discuss the modern methods to study CSCs in radiation biology, the role of CSCs in personalized cancer therapy as well as future directions for CSC research in translational radiooncology.
Collapse
Affiliation(s)
- Claudia Peitzsch
- a OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany.,b National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz-Zentrum Dresden - Rossendorf (HZDR) , Dresden , Germany.,c German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Ina Kurth
- d German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Nadja Ebert
- d German Cancer Research Center (DKFZ) , Heidelberg , Germany.,f Department of Radiotherapy and Radiation Oncology , Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany
| | - Anna Dubrovska
- a OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany.,c German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) , Heidelberg , Germany.,e Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay , Dresden , Germany
| | - Michael Baumann
- d German Cancer Research Center (DKFZ) , Heidelberg , Germany.,f Department of Radiotherapy and Radiation Oncology , Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
32
|
Barazzuol L, Hopkins SR, Ju L, Jeggo PA. Distinct response of adult neural stem cells to low versus high dose ionising radiation. DNA Repair (Amst) 2019; 76:70-75. [PMID: 30822688 DOI: 10.1016/j.dnarep.2019.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
Radiosusceptibility is the sensitivity of a biological organism to ionising radiation (IR)-induced carcinogenesis, an outcome of IR exposure relevant following low doses. The tissue response is strongly influenced by the DNA damage response (DDR) activated in stem and progenitor cells. We previously reported that in vivo exposure to 2 Gy X-rays activates apoptosis, proliferation arrest and premature differentiation in neural progenitor cells (transit amplifying cells and neuroblasts) but not in neural stem cells (NSCs) of the largest neurogenic region of the adult brain, the subventricular zone (SVZ). These responses promote adult quiescent NSC (qNSC) activation after 2 Gy. In contrast, neonatal (P5) SVZ neural progenitors continue proliferating and do not activate qNSCs. Significantly, the human and mouse neonatal brain is radiosusceptible. Here, we examine the response of stem and progenitor cells in the SVZ to low IR doses (50-500 mGy). We observe a linear dose-response for apoptosis but, in contrast, proliferation arrest and neuroblast differentiation require a threshold dose of 200 or 500 mGy, respectively. Importantly, qNSCs were not activated at doses below 500 mGy. Thus, full DDR activation in the neural stem cell compartment in vivo necessitates a threshold dose, which can be considered of significance when evaluating IR-induced cancer risk and dose extrapolation.
Collapse
Affiliation(s)
- Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, the Netherlands; Department of Biomedical Sciences of Cells and Systems, Section of Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, the Netherlands.
| | - Suzanna R Hopkins
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN1 9RQ, UK
| | - Limei Ju
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN1 9RQ, UK
| | - Penelope A Jeggo
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN1 9RQ, UK
| |
Collapse
|