1
|
Kuipers ME, van Liefferinge F, van der Wal E, Rovituso M, Slats AM, Hiemstra PS, Van Doorn-Wink KC. Effect of FLASH proton therapy on primary bronchial epithelial cell organoids. Clin Transl Radiat Oncol 2025; 52:100927. [PMID: 39968050 PMCID: PMC11833640 DOI: 10.1016/j.ctro.2025.100927] [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: 11/16/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Purpose The effects of conventional (CONV) and FLASH proton therapy on primary bronchial epithelial cell (PBEC) organoids from individuals with chronic obstructive pulmonary disease (COPD) were investigated. The primary objective was to compare the effect of FLASH and CONV on COPD PBEC organoids with a focus on DNA damage, organoid formation, and gene expression. Methods PBECs were obtained from six COPD donors, cultured as three-dimensional (3D) organoids and exposed to 2 and 8 Gy CONV and FLASH proton radiation at the Holland Proton Therapy Center. DNA damage was assessed by γH2AX staining. Organoid formation capacity was assessed by counting the organoids formed after reseeding irradiated cells at 24 h and 7 days. Bulk RNA sequencing (RNAseq) and qPCR analyses were performed to identify pathways and differences in the radiation response. Results γH2AX foci analysis showed a significant dose-dependent increase in DNA damage at 1 h for both CONV and FLASH treatments, without differences between the two modalities. Organoid formation assays revealed a dose-dependent decrease in organoid formation capacity at 24 h for both treatments. At 7 days, 2 Gy FLASH-treated samples showed significantly reduced organoid formation compared to 2 Gy CONV (p = 0.008). RNAseq identified CONV and FLASH-induced changes in expression of DNA-damage response and apoptosis pathway genes. A dose-dependent upregulation of MDM2, GDF15, DDB2, BAX, P21, AEN and a decrease in MKi67 expression was confirmed by qPCR analysis. Conclusion No significant differences were found in DNA damage or gene expression profiles between CONV and FLASH. The organoid formation assay showed a prolonged detrimental effect in the FLASH-treated organoids, suggesting a more complex interaction of FLASH with lung epithelial cells. The results of this study contribute to the advancement of robust in vitro human lung models for investigating the mechanisms of action of FLASH, potentially facilitating the treatment of NSCLC patients with proton FLASH therapy.
Collapse
Affiliation(s)
- Merian E. Kuipers
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Floriane van Liefferinge
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Ernst van der Wal
- Holland Proton Therapy Center (HollandPTC), Huismansingel 4 2629 JH Delft, the Netherlands
| | - Marta Rovituso
- Holland Proton Therapy Center (HollandPTC), Huismansingel 4 2629 JH Delft, the Netherlands
| | - Annelies M. Slats
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Pieter S. Hiemstra
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Krista C.J. Van Doorn-Wink
- Holland Proton Therapy Center (HollandPTC), Huismansingel 4 2629 JH Delft, the Netherlands
- Leiden University Medical Center (LUMC), Department of Radiotherapy, K01-P, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| |
Collapse
|
2
|
Akolawala Q, Accardo A. Engineered Cell Microenvironments: A Benchmark Tool for Radiobiology. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5563-5577. [PMID: 39813590 PMCID: PMC11788991 DOI: 10.1021/acsami.4c20455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
The development of engineered cell microenvironments for fundamental cell mechanobiology, in vitro disease modeling, and tissue engineering applications increased exponentially during the last two decades. In such context, in vitro radiobiology is a field of research aiming at understanding the effects of ionizing radiation (e.g., X-rays/photons, high-speed electrons, and high-speed protons) on biological (cancerous) tissues and cells, in particular in terms of DNA damage leading to cell death. Herein, the perspective provides a comparative assessment overview of scaffold-free, scaffold-based, and organ-on-a-chip models for radiobiology, highlighting opportunities, limitations, and future pathways to improve the currently existing approaches toward personalized cancer medicine.
Collapse
Affiliation(s)
- Qais Akolawala
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The
Netherlands
- Holland
Proton Therapy Center (HollandPTC), Huismansingel 4, 2629 JH Delft, The Netherlands
| | - Angelo Accardo
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The
Netherlands
| |
Collapse
|
3
|
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
|
4
|
Yu J, Wang K, Tang Y, Zheng D. Applications and perspectives of tumor organoids in radiobiology (Review). Oncol Rep 2024; 52:100. [PMID: 38904192 PMCID: PMC11223011 DOI: 10.3892/or.2024.8759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
Radiotherapy exhibits significant versatility and efficacy in cancer treatment, thereby playing a crucial role in the field of oncology. However, there remains an urgent need for extensive research on various aspects of radiotherapy, including target selection, damage repair and its combination with immunotherapy. Particularly, the development of in vitro models to replicate in vivo tumor lesion responses is vital. The present study provides a thorough review of the establishment and application of tumor organoids in radiotherapy, aiming to explore their potential impact on cancer treatment.
Collapse
Affiliation(s)
- Jin Yu
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Kailun Wang
- Emergency Department, Panshihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Yongjiang Tang
- Department of Vascular Surgery, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Dalin Zheng
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| |
Collapse
|
5
|
Reuvers TGA, Grandia V, Brandt RMC, Arab M, Maas SLN, Bos EM, Nonnekens J. Investigating the Radiobiological Response to Peptide Receptor Radionuclide Therapy Using Patient-Derived Meningioma Spheroids. Cancers (Basel) 2024; 16:2515. [PMID: 39061156 PMCID: PMC11275064 DOI: 10.3390/cancers16142515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using 177Lu-DOTA-TATE has recently been evaluated for the treatment of meningioma patients. However, current knowledge of the underlying radiation biology is limited, in part due to the lack of appropriate in vitro models. Here, we demonstrate proof-of-concept of a meningioma patient-derived 3D culture model to assess the short-term response to radiation therapies such as PRRT and external beam radiotherapy (EBRT). We established short-term cultures (1 week) for 16 meningiomas with high efficiency and yield. In general, meningioma spheroids retained characteristics of the parental tumor during the initial days of culturing. For a subset of tumors, clear changes towards a more aggressive phenotype were visible over time, indicating that the culture method induced dedifferentiation of meningioma cells. To assess PRRT efficacy, we demonstrated specific uptake of 177Lu-DOTA-TATE via somatostatin receptor subtype 2 (SSTR2), which was highly overexpressed in the majority of tumor samples. PRRT induced DNA damage which was detectable for an extended timeframe as compared to EBRT. Interestingly, levels of DNA damage in spheroids after PRRT correlated with SSTR2-expression levels of parental tumors. Our patient-derived meningioma culture model can be used to assess the short-term response to PRRT and EBRT in radiobiological studies. Further improvement of this model should pave the way towards the development of a relevant culture model for assessment of the long-term response to radiation and, potentially, individual patient responses to PRRT and EBRT.
Collapse
Affiliation(s)
- Thom G A Reuvers
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Vivian Grandia
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Majd Arab
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Sybren L N Maas
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Eelke M Bos
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Julie Nonnekens
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
6
|
Branciforti F, Salvi M, D’Agostino F, Marzola F, Cornacchia S, De Titta MO, Mastronuzzi G, Meloni I, Moschetta M, Porciani N, Sciscenti F, Spertini A, Spilla A, Zagaria I, Deloria AJ, Deng S, Haindl R, Szakacs G, Csiszar A, Liu M, Drexler W, Molinari F, Meiburger KM. Segmentation and Multi-Timepoint Tracking of 3D Cancer Organoids from Optical Coherence Tomography Images Using Deep Neural Networks. Diagnostics (Basel) 2024; 14:1217. [PMID: 38928633 PMCID: PMC11203156 DOI: 10.3390/diagnostics14121217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Recent years have ushered in a transformative era in in vitro modeling with the advent of organoids, three-dimensional structures derived from stem cells or patient tumor cells. Still, fully harnessing the potential of organoids requires advanced imaging technologies and analytical tools to quantitatively monitor organoid growth. Optical coherence tomography (OCT) is a promising imaging modality for organoid analysis due to its high-resolution, label-free, non-destructive, and real-time 3D imaging capabilities, but accurately identifying and quantifying organoids in OCT images remain challenging due to various factors. Here, we propose an automatic deep learning-based pipeline with convolutional neural networks that synergistically includes optimized preprocessing steps, the implementation of a state-of-the-art deep learning model, and ad-hoc postprocessing methods, showcasing good generalizability and tracking capabilities over an extended period of 13 days. The proposed tracking algorithm thoroughly documents organoid evolution, utilizing reference volumes, a dual branch analysis, key attribute evaluation, and probability scoring for match identification. The proposed comprehensive approach enables the accurate tracking of organoid growth and morphological changes over time, advancing organoid analysis and serving as a solid foundation for future studies for drug screening and tumor drug sensitivity detection based on organoids.
Collapse
Affiliation(s)
- Francesco Branciforti
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Massimo Salvi
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Filippo D’Agostino
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Francesco Marzola
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Sara Cornacchia
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Maria Olimpia De Titta
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Girolamo Mastronuzzi
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Isotta Meloni
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Miriam Moschetta
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Niccolò Porciani
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Fabrizio Sciscenti
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Alessandro Spertini
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Andrea Spilla
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Ilenia Zagaria
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Abigail J. Deloria
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Shiyu Deng
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Richard Haindl
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Gergely Szakacs
- Center for Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (G.S.); (A.C.)
| | - Agnes Csiszar
- Center for Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (G.S.); (A.C.)
| | - Mengyang Liu
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Wolfgang Drexler
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Filippo Molinari
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Kristen M. Meiburger
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| |
Collapse
|
7
|
Bouges E, Segers C, Leys N, Lebeer S, Zhang J, Mastroleo F. Human Intestinal Organoids and Microphysiological Systems for Modeling Radiotoxicity and Assessing Radioprotective Agents. Cancers (Basel) 2023; 15:5859. [PMID: 38136404 PMCID: PMC10741417 DOI: 10.3390/cancers15245859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Radiotherapy is a commonly employed treatment for colorectal cancer, yet its radiotoxicity-related impact on healthy tissues raises significant health concerns. This highlights the need to use radioprotective agents to mitigate these side effects. This review presents the current landscape of human translational radiobiology, outlining the limitations of existing models and proposing engineering solutions. We delve into radiotherapy principles, encompassing mechanisms of radiation-induced cell death and its influence on normal and cancerous colorectal cells. Furthermore, we explore the engineering aspects of microphysiological systems to represent radiotherapy-induced gastrointestinal toxicity and how to include the gut microbiota to study its role in treatment failure and success. This review ultimately highlights the main challenges and future pathways in translational research for pelvic radiotherapy-induced toxicity. This is achieved by developing a humanized in vitro model that mimics radiotherapy treatment conditions. An in vitro model should provide in-depth analyses of host-gut microbiota interactions and a deeper understanding of the underlying biological mechanisms of radioprotective food supplements. Additionally, it would be of great value if these models could produce high-throughput data using patient-derived samples to address the lack of human representability to complete clinical trials and improve patients' quality of life.
Collapse
Affiliation(s)
- Eloïse Bouges
- RadioPharma Research, Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.B.); (C.S.); (N.L.)
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium;
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands;
| | - Charlotte Segers
- RadioPharma Research, Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.B.); (C.S.); (N.L.)
| | - Natalie Leys
- RadioPharma Research, Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.B.); (C.S.); (N.L.)
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium;
| | - Jianbo Zhang
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands;
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, Location Academic Medical Center, 1105 BK Amsterdam, The Netherlands
| | - Felice Mastroleo
- RadioPharma Research, Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.B.); (C.S.); (N.L.)
| |
Collapse
|
8
|
de Nigris F, Meo C, Palinski W. Combination of Genomic Landsscape and 3D Culture Functional Assays Bridges Sarcoma Phenotype to Target and Immunotherapy. Cells 2023; 12:2204. [PMID: 37681936 PMCID: PMC10486752 DOI: 10.3390/cells12172204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
Genomic-based precision medicine has not only improved tumour therapy but has also shown its weaknesses. Genomic profiling and mutation analysis have identified alterations that play a major role in sarcoma pathogenesis and evolution. However, they have not been sufficient in predicting tumour vulnerability and advancing treatment. The relative rarity of sarcomas and the genetic heterogeneity between subtypes also stand in the way of gaining statistically significant results from clinical trials. Personalized three-dimensional tumour models that reflect the specific histologic subtype are emerging as functional assays to test anticancer drugs, complementing genomic screening. Here, we provide an overview of current target therapy for sarcomas and discuss functional assays based on 3D models that, by recapitulating the molecular pathways and tumour microenvironment, may predict patient response to treatments. This approach opens new avenues to improve precision medicine when genomic and pathway alterations are not sufficient to guide the choice of the most promising treatment. Furthermore, we discuss the aspects of the 3D culture assays that need to be improved, such as the standardisation of growth conditions and the definition of in vitro responses that can be used as a cut-off for clinical implementation.
Collapse
Affiliation(s)
- Filomena de Nigris
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Concetta Meo
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Wulf Palinski
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA;
| |
Collapse
|
9
|
Kang BK, Zhu Z, Wang J, Zhou J, Yu S, Zhou X, Zhao Z, Xie A, Lu L, Yang J. Maintenance of adult stem cells from human minor salivary glands via the Wnt signaling pathway. Stem Cell Res Ther 2023; 14:220. [PMID: 37620905 PMCID: PMC10464143 DOI: 10.1186/s13287-023-03445-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Xerostomia is a salivary gland dysfunction that negatively impacts the life quality of patients; however, there is no effective treatment for xerostomia. Bioengineered organs, generated using stem cells obtained from newborn salivary glands and ligated injury models, are a new organ transplantation strategy that could be feasible for xerostomia treatment. Reconstruction of salivary gland organoids by seed cells obtained from human minor salivary glands will offer theoretical fundaments and technology support for clinical application and organ regeneration research. Herein, we aimed to propose a new method for culturing and enriching adult human minor salivary gland stem cells in vitro in a three-dimensional (3D) environment via Wnt signaling activation. METHODS Obtained and characterized human minor salivary gland stem cells (hMSGSCs) with self-organization ability were 3D-cultured to generate organoids. We examined hMSGSCs proliferation and colony formation using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assays. Telomerase reverse transcriptase staining, flow cytometry, immunofluorescence assay, RNA isolation, RT-PCR, and qPCR were performed to assess hMSGSCs structure and the function of reconstructive organoids in vitro. RESULTS hMSGSCs showed typical epithelial-like characteristics, such as positive for CD49f and cell KRT expression. hMSGSCs served as adult stem cells in salivary glands and could differentiate into acinar and duct cells. Upon the addition of Noggin, CHIR99021, and Wnt3A to the 3D culture system, hMSGSCs showed higher LGR5 expression and decreased AMY1B and MUC5B expression. Therefore, the Wnt and bone morphogenetic protein (BMP) pathways are important in regulating hMSGSCs self-organization and differentiation. CONCLUSIONS We showed that the stem cell properties of hMSGSCs in a 3D culture system can be maintained by activating the Wnt signaling pathway and inhibiting the BMP signaling pathway. Our findings contribute new insights on salivary gland organoid generation in vitro.
Collapse
Affiliation(s)
- Bo Kyoung Kang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, China
| | - Zhu Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, China
| | - Jian Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shun Yu
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214041, China
| | - Xianyu Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhenmin Zhao
- Department of Plastic Surgery, Peking University 3Rd Hospital, NO.49 of North Huayuan Road, Haidian District, Beijing, 100191, China
| | - Aiguo Xie
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Lin Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
10
|
Functional precision oncology using patient-derived assays: bridging genotype and phenotype. Nat Rev Clin Oncol 2023; 20:305-317. [PMID: 36914745 DOI: 10.1038/s41571-023-00745-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 03/14/2023]
Abstract
Genomics-based precision medicine has revolutionized oncology but also has inherent limitations. Functional precision oncology is emerging as a complementary approach that aims to bridge the gap between genotype and phenotype by modelling individual tumours in vitro. These patient-derived ex vivo models largely preserve several tumour characteristics that are not captured by genomics approaches and enable the functional dissection of tumour vulnerabilities in a personalized manner. In this Review, we discuss several examples of personalized functional assays involving tumour organoids, spheroids and explants and their potential to predict treatment responses and drug-induced toxicities in individual patients. These developments have opened exciting new avenues for precision oncology, with the potential for successful clinical applications in contexts in which genomic data alone are not informative. To implement these assays into clinical practice, we outline four key barriers that need to be overcome: assay success rates, turnaround times, the need for standardized conditions and the definition of in vitro responders. Furthermore, we discuss novel technological advances such as microfluidics that might reduce sample requirements, assay times and labour intensity and thereby enable functional precision oncology to be implemented in routine clinical practice.
Collapse
|
11
|
Arjmand B, Rabbani Z, Soveyzi F, Tayanloo-Beik A, Rezaei-Tavirani M, Biglar M, Adibi H, Larijani B. Advancement of Organoid Technology in Regenerative Medicine. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023; 9:83-96. [PMID: 35968268 PMCID: PMC9360642 DOI: 10.1007/s40883-022-00271-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/25/2022]
Abstract
Purpose Organoids are three-dimensional cultures of stem cells in an environment similar to the body's extracellular matrix. This is also a novel development in the realm of regenerative medicine. Stem cells can begin to develop into 3D structures by modifying signaling pathways. To form organoids, stem cells are transplanted into the extracellular matrix. Organoids have provided the required technologies to reproduce human tissues. As a result, it might be used in place of animal models in scientific study. The key goals of these investigations are research into viral and genetic illnesses, malignancies, and extracellular vesicles, pharmaceutical discovery, and organ transplantation. Organoids can help pave the road for precision medicine through genetic editing, pharmaceutical development, and cell therapy. Methods PubMed, Google Scholar, and Scopus were used to search for all relevant papers written in English (1907-2021). The study abstracts were scrutinized. Studies on the use of stem-cell-derived organoids in regenerative medicine, organoids as 3D culture models for EVs analysis, and organoids for precision medicine were included. Articles with other irrelevant aims, meetings, letters, commentaries, congress and conference abstracts, and articles with no available full texts were excluded. Results According to the included studies, organoids have various origins, types, and applications in regenerative and precision medicine, as well as an important role in studying extracellular vesicles. Conclusion Organoids are considered a bridge that connects preclinical studies to clinical ones. However, the lack of a standardized protocol and other barriers addressed in this review, hinder the vast use of this technology. Lay Summary Organoids are 3D stem cell propagations in biological or synthetic scaffolds that mimic ECM to allow intercellular or matrix-cellular crosstalk. Because these structures are similar to organs in the body, they can be used as research models. Organoids are medicine's future hope for organ transplantation, tumor biobank formation, and the development of precision medicine. Organoid models can be used to study cell-to-cell interactions as well as effective factors like inflammation and aging. Bioengineering technologies are also used to define the size, shape, and composition of organoids before transforming them into precise structures. Finally, the importance of organoid applications in regenerative medicine has opened a new window for a better understanding of biological research, as discussed in this study.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Rabbani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Soveyzi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Adibi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Hong JA, Vikram B, Buchsbaum J, Capala J, Livinski A, Teicher B, Prasanna P, Ahmed MM, Obcemea C, Coleman CN, Espey MG. The State of Preclinical Modeling for Early Phase Cancer Trials Using Molecularly Targeted Agents with Radiation. Radiat Res 2022; 198:625-631. [PMID: 35976726 DOI: 10.1667/rade-22-00077.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/18/2022] [Indexed: 01/11/2023]
Abstract
Preclinical studies inform and guide the development of novel treatment combination strategies that bridge the laboratory with the clinic. We aimed to evaluate approaches cancer researchers used to justify advancing new combinations of molecularly targeted agents and radiation treatment into early-phase human clinical trials. Unsolicited early phase clinical trial proposals submitted to the National Cancer Institute's Cancer Therapy Evaluation Program between January 2016 and July 2020 were curated to quantify key characteristics and proportion of preclinical data provided by trialists seeking to conduct molecularly targeted agent-radiation combination studies in cancer patients. These data elucidate the current landscape for how the rationale for a molecularly targeted agent-radiation combination therapy is supported by preclinical research and illustrate unique challenges faced in translation at the intersection of precision medicine and radiation oncology.
Collapse
Affiliation(s)
- Julie A Hong
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - Bhadrasian Vikram
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - Jeffrey Buchsbaum
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | | | - Alicia Livinski
- National Institutes of Health Library, Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892
| | - Beverly Teicher
- Molecular Pharmacology Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - Pataje Prasanna
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - Mansoor M Ahmed
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - Ceferino Obcemea
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - C Norman Coleman
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| | - Michael Graham Espey
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland 20892
| |
Collapse
|
13
|
Radiotherapy Side Effects: Comprehensive Proteomic Study Unraveled Neural Stem Cell Degenerative Differentiation upon Ionizing Radiation. Biomolecules 2022; 12:biom12121759. [PMID: 36551187 PMCID: PMC9775306 DOI: 10.3390/biom12121759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Cranial radiation therapy is one of the most effective treatments for childhood brain cancers. Despite the ameliorated survival rate of juvenile patients, radiation exposure-induced brain neurogenic region injury could markedly impair patients' cognitive functions and even their quality of life. Determining the mechanism underlying neural stem cells (NSCs) response to irradiation stress is a crucial therapeutic strategy for cognitive impairment. The present study demonstrated that X-ray irradiation arrested NSCs' cell cycle and impacted cell differentiation. To further characterize irradiation-induced molecular alterations in NSCs, two-dimensional high-resolution mass spectrometry-based quantitative proteomics analyses were conducted to explore the mechanism underlying ionizing radiation's influence on stem cell differentiation. We observed that ionizing radiation suppressed intracellular protein transport, neuron projection development, etc., particularly in differentiated cells. Redox proteomics was performed for the quantification of cysteine thiol modifications in order to profile the oxidation-reduction status of proteins in stem cells that underwent ionizing radiation treatment. Via conjoint screening of protein expression abundance and redox status datasets, several significantly expressed and oxidized proteins were identified in differentiating NSCs subjected to X-ray irradiation. Among these proteins, succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial (sdha) and the acyl carrier protein, mitochondrial (Ndufab1) were highly related to neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and Huntington's disease, illustrating the dual-character of NSCs in cell differentiation: following exposure to ionizing radiation, the normal differentiation of NSCs was compromised, and the upregulated oxidized proteins implied a degenerative differentiation trajectory. These findings could be integrated into research on neurodegenerative diseases and future preventive strategies.
Collapse
|
14
|
Naumann M, Czempiel T, Lößner AJ, Pape K, Beyreuther E, Löck S, Drukewitz S, Hennig A, von Neubeck C, Klink B, Krause M, William D, Stange DE, Bütof R, Dietrich A. Combined Systemic Drug Treatment with Proton Therapy: Investigations on Patient-Derived Organoids. Cancers (Basel) 2022; 14:cancers14153781. [PMID: 35954444 PMCID: PMC9367296 DOI: 10.3390/cancers14153781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
To optimize neoadjuvant radiochemotherapy of pancreatic ductal adenocarcinoma (PDAC), the value of new irradiation modalities such as proton therapy needs to be investigated in relevant preclinical models. We studied individual treatment responses to RCT using patient-derived PDAC organoids (PDO). Four PDO lines were treated with gemcitabine, 5-fluorouracile (5FU), photon and proton irradiation and combined RCT. Therapy response was subsequently measured via viability assays. In addition, treatment-naive PDOs were characterized via whole exome sequencing and tumorigenicity was investigated in NMRI Foxn1nu/nu mice. We found a mutational pattern containing common mutations associated with PDAC within the PDOs. Although we could unravel potential complications of the viability assay for PDOs in radiobiology, distinct synergistic effects of gemcitabine and 5FU with proton irradiation were observed in two PDO lines that may lead to further mechanistical studies. We could demonstrate that PDOs are a powerful tool for translational proton radiation research.
Collapse
Affiliation(s)
- Max Naumann
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Tabea Czempiel
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (T.C.); (S.D.); (B.K.); (D.W.)
| | - Anna Jana Lößner
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (A.J.L.); (K.P.); (A.H.); (D.E.S.)
| | - Kristin Pape
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (A.J.L.); (K.P.); (A.H.); (D.E.S.)
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- Institute of Radiation Physics, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Steffen Löck
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany
| | - Stephan Drukewitz
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (T.C.); (S.D.); (B.K.); (D.W.)
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Alexander Hennig
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (A.J.L.); (K.P.); (A.H.); (D.E.S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Cläre von Neubeck
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany
- Clinic for Particle Therapy, University Hospital Essen, Universität Duisburg Essen, 45147 Essen, Germany
| | - Barbara Klink
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (T.C.); (S.D.); (B.K.); (D.W.)
- Department of Genetics, Laboratoire National de Santé, 3555 Dudelange, Luxembourg
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus, Technische Universität Dresden, ERN-GENTURIS, Hereditary Cancer Syndrome Center Dresden, 01307 Dresden, Germany
| | - Mechthild Krause
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany
| | - Doreen William
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (T.C.); (S.D.); (B.K.); (D.W.)
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus, Technische Universität Dresden, ERN-GENTURIS, Hereditary Cancer Syndrome Center Dresden, 01307 Dresden, Germany
| | - Daniel E. Stange
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (A.J.L.); (K.P.); (A.H.); (D.E.S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Rebecca Bütof
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany
| | - Antje Dietrich
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (M.N.); (E.B.); (S.L.); (C.v.N.); (M.K.); (R.B.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
15
|
Orzechowska-Licari EJ, LaComb JF, Giarrizzo M, Yang VW, Bialkowska AB. Intestinal Epithelial Regeneration in Response to Ionizing Irradiation. J Vis Exp 2022:10.3791/64028. [PMID: 35969101 PMCID: PMC9631267 DOI: 10.3791/64028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
The intestinal epithelium consists of a single layer of cells yet contains multiple types of terminally differentiated cells, which are generated by the active proliferation of intestinal stem cells located at the bottom of intestinal crypts. However, during events of acute intestinal injury, these active intestinal stem cells undergo cell death. Gamma irradiation is a widely used colorectal cancer treatment, which, while therapeutically efficacious, has the side effect of depleting the active stem cell pool. Indeed, patients frequently experience gastrointestinal radiation syndrome while undergoing radiotherapy, in part due to active stem cell depletion. The loss of active intestinal stem cells in intestinal crypts activates a pool of typically quiescent reserve intestinal stem cells and induces dedifferentiation of secretory and enterocyte precursor cells. If not for these cells, the intestinal epithelium would lack the ability to recover from radiotherapy and other such major tissue insults. New advances in lineage-tracing technologies allow tracking of the activation, differentiation, and migration of cells during regeneration and have been successfully employed for studying this in the gut. This study aims to depict a method for the analysis of cells within the mouse intestinal epithelium following radiation injury.
Collapse
Affiliation(s)
| | - Joseph F LaComb
- Department of Medicine, Renaissance School of Medicine at Stony Brook University
| | - Michael Giarrizzo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University
| | - Vincent W Yang
- Department of Medicine, Renaissance School of Medicine at Stony Brook University; Department of Physiology and Biophysics, Renaissance School of Medicine at Stony Brook University
| | | |
Collapse
|
16
|
Forsythe SD, Sivakumar H, Erali RA, Wajih N, Li W, Shen P, Levine EA, Miller KE, Skardal A, Votanopoulos KI. Patient-Specific Sarcoma Organoids for Personalized Translational Research: Unification of the Operating Room with Rare Cancer Research and Clinical Implications. Ann Surg Oncol 2022; 29:7354-7367. [PMID: 35780216 DOI: 10.1245/s10434-022-12086-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/10/2022] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Sarcoma clinical outcomes have been stagnant for decades due to heterogeneity of primaries, lack of comprehensive preclinical models, and rarity of disease. We hypothesized that engineering hydrogel-based sarcoma organoids directly from the patient without xenogeneic extracellular matrices (ECMs) or growth factors is routinely feasible and allows rare tumors to remain viable as avatars for personalized research. METHODS Surgically resected sarcomas (angiosarcomas, leiomyosarcoma, gastrointestinal stromal tumor, liposarcoma, myxofibrosarcoma, dermatofibrosarcoma protuberans [DFSP], and pleiomorphic abdominal sarcoma) were dissociated and incorporated into a hyaluronic acid and collagen-based ECM hydrogel and screened for chemotherapy efficacy. A subset of organoids was enriched with a patient-matched immune system for screening of immunotherapy efficacy (iPTOs). Response to treatment was assessed using LIVE/DEAD staining and metabolic assays. RESULTS Sixteen sarcomas were biofabricated into three-dimensional (3D) patient-specific sarcoma organoids with a 100% success rate. Average time from organoid development to initiation of drug testing was 7 days. Enrichment of organoids with immune system components derived from either peripheral blood mononuclear cells or lymph node cells was performed in 10/16 (62.5%) patients; 4/12 (33%) organoids did not respond to chemotherapy, while response to immunotherapy was observed in 2/10 (20%) iPTOs. CONCLUSIONS A large subset of sarcoma organoids does not exhibit response to chemotherapy or immunotherapy, as currently seen in clinical practice. Routine development of sarcoma hydrogel-based organoids directly from the operating room is a feasible platform, allowing for such rare tumors to remain viable for personalized translational research.
Collapse
Affiliation(s)
- Steven D Forsythe
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Organoid Research Center (WFORCE), Winston-Salem, NC, USA
| | - Hemamylammal Sivakumar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Richard A Erali
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Organoid Research Center (WFORCE), Winston-Salem, NC, USA.,Department of Surgery, Division of Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nadeem Wajih
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Organoid Research Center (WFORCE), Winston-Salem, NC, USA
| | - Wencheng Li
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Perry Shen
- Department of Surgery, Division of Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Edward A Levine
- Department of Surgery, Division of Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Katherine E Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA. .,The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA.
| | - Konstantinos I Votanopoulos
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Wake Forest Organoid Research Center (WFORCE), Winston-Salem, NC, USA. .,Department of Surgery, Division of Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA. .,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
17
|
Wang Y, Li Y, Sheng Z, Deng W, Yuan H, Wang S, Liu Y. Advances of Patient-Derived Organoids in Personalized Radiotherapy. Front Oncol 2022; 12:888416. [PMID: 35574360 PMCID: PMC9102799 DOI: 10.3389/fonc.2022.888416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/28/2022] [Indexed: 11/16/2022] Open
Abstract
Patient-derived organoids (PDO), based on the advanced three-dimensional (3D) culture technology, can provide more relevant physiological and pathological cancer models, which is especially beneficial for developing and optimizing cancer therapeutic strategies. Radiotherapy (RT) is a cornerstone of curative and palliative cancer treatment, which can be performed alone or integrated with surgery, chemotherapy, immunotherapy, or targeted therapy in clinical care. Among all cancer therapies, RT has great local control, safety and effectiveness, and is also cost-effective per life-year gained for patients. It has been reported that combing RT with chemotherapy or immunotherapy or radiosensitizer drugs may enhance treatment efficacy at faster rates and lower cost. However, very few FDA-approved combinations of RT with drugs or radiosensitizers exist due to the lack of accurate and relevant preclinical models. Meanwhile, radiation dose escalation may increase treatment efficacy and induce more toxicity of normal tissue as well, which has been studied by conducting various clinical trials, very expensive and time-consuming, often burdensome on patients and sometimes with controversial results. The surged PDO technology may help with the preclinical test of RT combination and radiation dose escalation to promote precision radiation oncology, where PDO can recapitulate individual patient’ tumor heterogeneity, retain characteristics of the original tumor, and predict treatment response. This review aims to introduce recent advances in the PDO technology and personalized radiotherapy, highlight the strengths and weaknesses of the PDO cancer models, and finally examine the existing RT-related PDO trials or applications to harness personalized and precision radiotherapy.
Collapse
Affiliation(s)
- Yuenan Wang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Yuenan Wang, ; Yajie Liu, ; Shubin Wang,
| | - Ye Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weiwei Deng
- Department of Mechanical and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Hongyan Yuan
- Department of Mechanical and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Shubin Wang
- Department of Medical Oncology, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Yuenan Wang, ; Yajie Liu, ; Shubin Wang,
| | - Yajie Liu
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Yuenan Wang, ; Yajie Liu, ; Shubin Wang,
| |
Collapse
|
18
|
Lehmann V, Schene IF, Ardisasmita AI, Liv N, Veenendaal T, Klumperman J, van der Doef HPJ, Verkade HJ, Verstegen MMA, van der Laan LJW, Jans JJM, Verhoeven‐Duif NM, van Hasselt PM, Nieuwenhuis EES, Spee B, Fuchs SA. The potential and limitations of intrahepatic cholangiocyte organoids to study inborn errors of metabolism. J Inherit Metab Dis 2022; 45:353-365. [PMID: 34671987 PMCID: PMC9298016 DOI: 10.1002/jimd.12450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 01/09/2023]
Abstract
Inborn errors of metabolism (IEMs) comprise a diverse group of individually rare monogenic disorders that affect metabolic pathways. Mutations lead to enzymatic deficiency or dysfunction, which results in intermediate metabolite accumulation or deficit leading to disease phenotypes. Currently, treatment options for many IEMs are insufficient. Rarity of individual IEMs hampers therapy development and phenotypic and genetic heterogeneity suggest beneficial effects of personalized approaches. Recently, cultures of patient-own liver-derived intrahepatic cholangiocyte organoids (ICOs) have been established. Since most metabolic genes are expressed in the liver, patient-derived ICOs represent exciting possibilities for in vitro modeling and personalized drug testing for IEMs. However, the exact application range of ICOs remains unclear. To address this, we examined which metabolic pathways can be studied with ICOs and what the potential and limitations of patient-derived ICOs are to model metabolic functions. We present functional assays in patient ICOs with defects in branched-chain amino acid metabolism (methylmalonic acidemia), copper metabolism (Wilson disease), and transporter defects (cystic fibrosis). We discuss the broad range of functional assays that can be applied to ICOs, but also address the limitations of these patient-specific cell models. In doing so, we aim to guide the selection of the appropriate cell model for studies of a specific disease or metabolic process.
Collapse
Affiliation(s)
- Vivian Lehmann
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Imre F. Schene
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Arif I. Ardisasmita
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Nalan Liv
- Section Cell Biology, Center for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Tineke Veenendaal
- Section Cell Biology, Center for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Henkjan J. Verkade
- Department of Pediatric GastroenterologyUniversity Medical Center GroningenGroningenThe Netherlands
- Department of HepatologyUniversity Medical Center GroningenGroningenThe Netherlands
| | | | | | - Judith J. M. Jans
- Department of Metabolic DiagnosticsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Peter M. van Hasselt
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Bart Spee
- Department of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Sabine A. Fuchs
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
19
|
Overgaard J, Aznar MC, Bacchus C, Coppes RP, Deutsch E, Georg D, Haustermans K, Hoskin P, Krause M, Lartigau EF, Lee AWM, Löck S, Offersen BV, Thwaites DI, van der Kogel AJ, van der Heide UA, Valentini V, Baumann M. Personalised radiation therapy taking both the tumour and patient into consideration. Radiother Oncol 2022; 166:A1-A5. [PMID: 35051440 DOI: 10.1016/j.radonc.2022.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark.
| | - Marianne C Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, The Christie NHS Foundation Trust, United Kingdom
| | - Carol Bacchus
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rob P Coppes
- Departments of Radiation Oncology and Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Eric Deutsch
- Department of Radiation Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, France
| | - Dietmar Georg
- Division Medical Radiation Physics, Department of Radiation Oncology, Medical University of Vienna/AKH Wien, Austria
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Belgium
| | - Peter Hoskin
- Mount Vernon Cancer Centre and University of Manchester, United Kingdom
| | - Mechthild Krause
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany
| | - Eric F Lartigau
- Academic Department of Radiotherapy, Oscar Lambret Comprehensive Cancer Center, Lille, France
| | - Anne W M Lee
- Department of Clinical Oncology, University of Hong Kong - Shenzhen Hospital and University of Hong Kong, China
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany
| | - Birgitte V Offersen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark
| | - David I Thwaites
- Institute of Medical Physics, School of Physics, The University of Sydney, Australia; Medical Physics Group, Leeds Institute of Medical Research, School of Medicine, University of Leeds, United Kingdom
| | - Albert J van der Kogel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Uulke A van der Heide
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vincenzo Valentini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, UOC Radioterapia Oncologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | | |
Collapse
|
20
|
Suckert T, Nexhipi S, Dietrich A, Koch R, Kunz-Schughart LA, Bahn E, Beyreuther E. Models for Translational Proton Radiobiology-From Bench to Bedside and Back. Cancers (Basel) 2021; 13:4216. [PMID: 34439370 PMCID: PMC8395028 DOI: 10.3390/cancers13164216] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022] Open
Abstract
The number of proton therapy centers worldwide are increasing steadily, with more than two million cancer patients treated so far. Despite this development, pending questions on proton radiobiology still call for basic and translational preclinical research. Open issues are the on-going discussion on an energy-dependent varying proton RBE (relative biological effectiveness), a better characterization of normal tissue side effects and combination treatments with drugs originally developed for photon therapy. At the same time, novel possibilities arise, such as radioimmunotherapy, and new proton therapy schemata, such as FLASH irradiation and proton mini-beams. The study of those aspects demands for radiobiological models at different stages along the translational chain, allowing the investigation of mechanisms from the molecular level to whole organisms. Focusing on the challenges and specifics of proton research, this review summarizes the different available models, ranging from in vitro systems to animal studies of increasing complexity as well as complementing in silico approaches.
Collapse
Affiliation(s)
- Theresa Suckert
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sindi Nexhipi
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01309 Dresden, Germany
| | - Antje Dietrich
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Robin Koch
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Leoni A. Kunz-Schughart
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Emanuel Bahn
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, 69120 Heidelberg, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiation Physics, 01328 Dresden, Germany
| |
Collapse
|
21
|
Stockton JD, Tee L, Whalley C, James J, Dilworth M, Wheat R, Nieto T, Geh I, Barros-Silva JD, Beggs AD. Complete response to neoadjuvant chemoradiotherapy in rectal cancer is associated with RAS/AKT mutations and high tumour mutational burden. Radiat Oncol 2021; 16:129. [PMID: 34256782 PMCID: PMC8278688 DOI: 10.1186/s13014-021-01853-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pathological complete response (pathCR) in rectal cancer is beneficial, as up to 75% of patients do not experience regrowth of the primary tumour, but it is poorly understood. We hypothesised that the changes seen in the pre-treatment biopsies of pathCR but not seen in residual tumour after chemoradiotherapy were the determinants of responsiveness. METHODS Two groups of patients with either complete response (pathCR group, N = 24) or no response (poor response group, N = 24) were retrieved. Pre-treatment biopsies of cancers from these patients underwent high read depth amplicon sequencing for a targeted panel, exome sequencing, methylation profiling and immunohistochemistry for DNA repair pathway proteins. RESULTS Twenty four patients who underwent pathCR and twenty-four who underwent poor response underwent molecular characterisation. Patients in the pathCR group had significantly higher tumour mutational burden and neoantigen load, frequent copy number alterations but fewer structural variants and enrichment for driver mutations in the PI3K/AKT/mTOR signalling pathway. There were no significant differences in tumour heterogeneity as measured by MATH score. Methylation analysis demonstrated enrichment for hypomethyation in the PI3K/AKT/mTOR signalling pathway. DISCUSSION The phenomenon of pathCR in rectal cancer may be related to immunovisibility caused by a high tumour mutational burden phenotype. Potential therapy resistance mechanisms involve the PI3K/AKT/mTOR signalling pathway, but tumour heterogeneity does not seem to play a role in resistance.
Collapse
Affiliation(s)
- Joanne D. Stockton
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Louise Tee
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Celina Whalley
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Jonathan James
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Mark Dilworth
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Rachel Wheat
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Thomas Nieto
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - S-CORT Consortium
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Ian Geh
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - João D. Barros-Silva
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Andrew D. Beggs
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
22
|
|