1
|
Wiebe M, Milligan K, Brewer J, Fuentes AM, Ali-Adeeb R, Brolo AG, Lum JJ, Andrews JL, Haston C, Jirasek A. Metabolic profiling of murine radiation-induced lung injury with Raman spectroscopy and comparative machine learning. Analyst 2024; 149:2864-2876. [PMID: 38619825 DOI: 10.1039/d4an00152d] [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: 04/16/2024]
Abstract
Radiation-induced lung injury (RILI) is a dose-limiting toxicity for cancer patients receiving thoracic radiotherapy. As such, it is important to characterize metabolic associations with the early and late stages of RILI, namely pneumonitis and pulmonary fibrosis. Recently, Raman spectroscopy has shown utility for the differentiation of pneumonitic and fibrotic tissue states in a mouse model; however, the specific metabolite-disease associations remain relatively unexplored from a Raman perspective. This work harnesses Raman spectroscopy and supervised machine learning to investigate metabolic associations with radiation pneumonitis and pulmonary fibrosis in a mouse model. To this end, Raman spectra were collected from lung tissues of irradiated/non-irradiated C3H/HeJ and C57BL/6J mice and labelled as normal, pneumonitis, or fibrosis, based on histological assessment. Spectra were decomposed into metabolic scores via group and basis restricted non-negative matrix factorization, classified with random forest (GBR-NMF-RF), and metabolites predictive of RILI were identified. To provide comparative context, spectra were decomposed and classified via principal component analysis with random forest (PCA-RF), and full spectra were classified with a convolutional neural network (CNN), as well as logistic regression (LR). Through leave-one-mouse-out cross-validation, we observed that GBR-NMF-RF was comparable to other methods by measure of accuracy and log-loss (p > 0.10 by Mann-Whitney U test), and no methodology was dominant across all classification tasks by measure of area under the receiver operating characteristic curve. Moreover, GBR-NMF-RF results were directly interpretable and identified collagen and specific collagen precursors as top fibrosis predictors, while metabolites with immune and inflammatory functions, such as serine and histidine, were top pneumonitis predictors. Further support for GBR-NMF-RF and the identified metabolite associations with RILI was found as CNN interpretation heatmaps revealed spectral regions consistent with these metabolites.
Collapse
Affiliation(s)
- Mitchell Wiebe
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Kirsty Milligan
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Joan Brewer
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Alejandra M Fuentes
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Ramie Ali-Adeeb
- Department of Chemistry, The University of Victoria, Victoria, Canada
| | - Alexandre G Brolo
- Department of Chemistry, The University of Victoria, Victoria, Canada
| | - Julian J Lum
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, Canada
| | - Jeffrey L Andrews
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Christina Haston
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Andrew Jirasek
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| |
Collapse
|
2
|
Chen Q, Guo X, Wang H, Sun S, Jiang H, Zhang P, Shang E, Zhang R, Cao Z, Niu Q, Zhang C, Liu Y, Shi L, Yu Y, Hou W, Zheng Y. Plasma-Free Blood as a Potential Alternative to Whole Blood for Transcriptomic Analysis. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:109-124. [PMID: 38884056 PMCID: PMC11169349 DOI: 10.1007/s43657-023-00121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/29/2023] [Accepted: 07/13/2023] [Indexed: 06/18/2024]
Abstract
RNA sequencing (RNAseq) technology has become increasingly important in precision medicine and clinical diagnostics, and emerged as a powerful tool for identifying protein-coding genes, performing differential gene analysis, and inferring immune cell composition. Human peripheral blood samples are widely used for RNAseq, providing valuable insights into individual biomolecular information. Blood samples can be classified as whole blood (WB), plasma, serum, and remaining sediment samples, including plasma-free blood (PFB) and serum-free blood (SFB) samples that are generally considered less useful byproducts during the processes of plasma and serum separation, respectively. However, the feasibility of using PFB and SFB samples for transcriptome analysis remains unclear. In this study, we aimed to assess the suitability of employing PFB or SFB samples as an alternative RNA source in transcriptomic analysis. We performed a comparative analysis of WB, PFB, and SFB samples for different applications. Our results revealed that PFB samples exhibit greater similarity to WB samples than SFB samples in terms of protein-coding gene expression patterns, detection of differentially expressed genes, and immunological characterizations, suggesting that PFB can serve as a viable alternative to WB for transcriptomic analysis. Our study contributes to the optimization of blood sample utilization and the advancement of precision medicine research. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-023-00121-1.
Collapse
Affiliation(s)
- Qingwang Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Xiaorou Guo
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Haiyan Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Shanyue Sun
- Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021 China
| | - He Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Peipei Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Erfei Shang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Ruolan Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Zehui Cao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Quanne Niu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Chao Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Yaqing Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
- The International Human Phenome Institutes, Shanghai, 200438 China
| | - Ying Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Wanwan Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Yuanting Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| |
Collapse
|
3
|
Shakyawar SK, Mishra NK, Vellichirammal NN, Cary L, Helikar T, Powers R, Oberley-Deegan RE, Berkowitz DB, Bayles KW, Singh VK, Guda C. A Review of Radiation-Induced Alterations of Multi-Omic Profiles, Radiation Injury Biomarkers, and Countermeasures. Radiat Res 2023; 199:89-111. [PMID: 36368026 PMCID: PMC10279411 DOI: 10.1667/rade-21-00187.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
Increasing utilization of nuclear power enhances the risks associated with industrial accidents, occupational hazards, and the threat of nuclear terrorism. Exposure to ionizing radiation interferes with genomic stability and gene expression resulting in the disruption of normal metabolic processes in cells and organs by inducing complex biological responses. Exposure to high-dose radiation causes acute radiation syndrome, which leads to hematopoietic, gastrointestinal, cerebrovascular, and many other organ-specific injuries. Altered genomic variations, gene expression, metabolite concentrations, and microbiota profiles in blood plasma or tissue samples reflect the whole-body radiation injuries. Hence, multi-omic profiles obtained from high-resolution omics platforms offer a holistic approach for identifying reliable biomarkers to predict the radiation injury of organs and tissues resulting from radiation exposures. In this review, we performed a literature search to systematically catalog the radiation-induced alterations from multi-omic studies and radiation countermeasures. We covered radiation-induced changes in the genomic, transcriptomic, proteomic, metabolomic, lipidomic, and microbiome profiles. Furthermore, we have covered promising multi-omic biomarkers, FDA-approved countermeasure drugs, and other radiation countermeasures that include radioprotectors and radiomitigators. This review presents an overview of radiation-induced alterations of multi-omics profiles and biomarkers, and associated radiation countermeasures.
Collapse
Affiliation(s)
- Sushil K Shakyawar
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nitish K Mishra
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Neetha N Vellichirammal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lynnette Cary
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln NE 65888, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln NE 65888, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln NE 68588, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David B Berkowitz
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln NE 65888, USA
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
4
|
Bucknell NW, Belderbos J, Palma DA, Iyengar P, Samson P, Chua K, Gomez D, McDonald F, Louie AV, Faivre-Finn C, Hanna GG, Siva S. Avoiding toxicity with lung radiation therapy: An IASLC perspective. J Thorac Oncol 2022; 17:961-973. [DOI: 10.1016/j.jtho.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022]
|
5
|
Gkika E, Adebahr S, Brenner A, Schimek-Jasch T, Radicioni G, Exner JP, Rühle A, Spohn SKB, Popp I, Zamboglou C, Sprave T, Firat E, Niedermann G, Nicolay NH, Nestle U, Grosu AL, Duda DG. Changes in Blood Biomarkers of Angiogenesis and Immune Modulation after Radiation Therapy and Their Association with Outcomes in Thoracic Malignancies. Cancers (Basel) 2021; 13:cancers13225725. [PMID: 34830880 PMCID: PMC8616228 DOI: 10.3390/cancers13225725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
The effects of radiotherapy on systemic immunity remain to be fully characterized in a disease-specific manner. The aim of the study was to examine potential biomarkers of systemic immunomodulation when using radiotherapy for thoracic malignancies. Serial blood samples were collected from 56 patients with thoracic malignancies prior (RTbaseline), during (RTduring) and at the end of radiotherapy (RTend), as well as at the first (FU1) and second follow-up (FU2). The changes in serum levels of IL-10, IFN-γ, IL-12p70, IL-13, IL-1β, IL-4, IL-6, IL-8, TNF-α, bFGF, sFlt-1, PlGF, VEGF, VEGF-C, VEGF-D and HGF were measured by multiplexed array and tested for associations with clinical outcomes. We observed an increase in the levels of IL-10, IFN-γ, PlGF and VEGF-D and a decrease in those of IL-8, VEGF, VEGF-C and sFlt-1 during and at the end of radiotherapy. Furthermore, baseline concentration of TNF-α significantly correlated with OS. IL-6 level at RTend and FU1,2 correlated with OS (RTend: p = 0.039, HR: 1.041, 95% CI: 1.002-1.082, FU1: p = 0.001, HR: 1.139, 95% CI: 1.056-1.228, FU2: p = 0.017, HR: 1.101 95% CI: 1.018-1.192), while IL-8 level correlated with OS at RTduring and RTend (RTduring: p = 0.017, HR: 1.014, 95% CI: 1.002-1.026, RTend: p = 0.004, HR: 1.007, 95% CI: 1.061-1.686). In conclusion, serum levels of TNF-α, IL-6 and IL-8 are potential biomarkers of response to radiotherapy. Given the recent implementation of immunotherapy in lung and esophageal cancer, these putative blood biomarkers should be further validated and evaluated in the combination or sequential therapy setting.
Collapse
Affiliation(s)
- Eleni Gkika
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| | - Sonja Adebahr
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anton Brenner
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
| | - Tanja Schimek-Jasch
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gianluca Radicioni
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jan-Philipp Exner
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Rühle
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Simon K. B. Spohn
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ilinca Popp
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Constantinos Zamboglou
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Tanja Sprave
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Elke Firat
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gabriele Niedermann
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Nils Henrik Nicolay
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ursula Nestle
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- Department of Radiation Oncology, Kliniken Maria Hilf, 41063 Moenchengladbach, Germany
| | - Anca-Ligia Grosu
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dan G. Duda
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| |
Collapse
|
6
|
Deek MP, Yegya-Raman N, Daroui P, Balasubramanian S, Malhotra J, Moore D, Patel M, Wang SJ, Aisner J, Jabbour SK. Overexpression of excision repair cross-complementing 1 gene associates with higher risk of therapeutic failure after definitive chemoradiation for unresectable non-small cell lung cancer. ANNALS OF PALLIATIVE MEDICINE 2021; 10:7205-7213. [PMID: 34263627 PMCID: PMC10845888 DOI: 10.21037/apm-21-182] [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: 01/22/2021] [Accepted: 05/18/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Locally advanced non-small cell lung cancer (NSCLC) is typically treated with concurrent chemoradiation (CRT). Excision Repair Cross-Complementing 1 (ERCC1) is a protein involved in DNA damage repair. The objective of this study was to assess whether higher tumoral ERCC1 expression would associate with worse clinical outcomes in NSCLC treated with CRT. METHODS Twenty-five patients were included. Relative expression levels of messenger RNA (mRNA) for ERCC1 were measured with a quantitative reverse transcription polymerase chain reaction (qRT-PCR) and expressed as scaled ERCC1 mRNA gene expression value. Patients were followed every 3 months with history, physical exam, and imaging to assess clinical outcomes. We evaluated the associations between ERCC1, as well as other prognostic variables including stage, age, gender, race, histology, RT dose, performance status, and progression free survival (PFS) and overall survival (OS) with Kaplan-Meier method and Cox regression. RESULTS Recursive partitioning analysis identified a GeneExp cutoff of 1.54. Higher ERCC1 expression was associated with worse PFS [hazard ratio (HR) =1.70, P=0.04] and trended towards worse OS (HR =1.53, P=0.11). Increasing tumor volume (HR =1.001, P=0.055), squamous cell (HR =7.86, P=0.008) and poorly differentiated histology (HR =5.25, P=0.06) also associated with worse OS. The cumulative incidence of local recurrence at 1 year trended higher with ERCC1 GeneExp ≥1.54 (78.1%) compared to ERCC1 GeneExp <1.54 (14.9%, P=0.08). Distant relapse at 1 year was 72% with tumor ERCC1 expression ≥1.54 and 52% with ERCC1 expression <1.54 (P=0.28). CONCLUSIONS Higher ERCC1 expression by qRT-PCR appears to correlate with worse PFS in locally advanced NSCLC treated with CRT. However, the overall sample size of the population was small; thus, larger studies are warranted to integrate molecular biomarkers to identify patients who might benefit from treatment intensification.
Collapse
Affiliation(s)
- Matthew P. Deek
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nikhil Yegya-Raman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Parima Daroui
- Department of Radiation Oncology, The Permanente Medical Group, Santa Clara Cancer Treatment Center, Santa Clara, CA, USA
| | | | - Jyoti Malhotra
- Department of Medical Oncology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Dirk Moore
- Department of Biostatistics and Epidemiology, Rutgers University, NJ, USA
| | - Malini Patel
- Department of Medical Oncology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Shang-Jui Wang
- Department of Radiation Oncology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Joseph Aisner
- Department of Medical Oncology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Salma K. Jabbour
- Department of Radiation Oncology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| |
Collapse
|
7
|
Sui X, Jiang L, Teng H, Mi L, Li B, Shi A, Yu R, Li D, Dong X, Yang D, Yu H, Wang W. Prediction of Clinical Outcome in Locally Advanced Non-Small Cell Lung Cancer Patients Treated With Chemoradiotherapy by Plasma Markers. Front Oncol 2021; 10:625911. [PMID: 33680949 PMCID: PMC7925829 DOI: 10.3389/fonc.2020.625911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/30/2020] [Indexed: 12/28/2022] Open
Abstract
Purpose To identify cytokines in plasma that may predict objective response and progression-free survival (PFS) in patients with locally advanced non-small cell lung cancer (NSCLC) treated with chemoradiotherapy. Materials and Methods From April 2016 to May 2017, thirty-one patients with locally advanced inoperable/unresectable NSCLC were included, and treated with concurrent chemoradiotherapy (CCRT). No immune checkpoint inhibitors were administered after CCRT. Plasma from each patient was collected before radiotherapy, and 25 cytokines in the plasma were measured by Luminex or U-PLEX assays. Logistic regression and COX regression were performed to identify the predictive factors for objective response and PFS, respectively. Kaplan-Meier survival analysis was used to compare the PFS between the groups. Results High levels of IL-13 and TNF-α, and low levels of ICAM-1, IFN-γ, and soluble PD-L1 (sPD-L1) were significantly associated with objective response (P <0.05). High levels of IL-8, CCL5, and CXCL3 also showed a trend toward association with objective response (P <0.1). The combination of cytokines (IL-8 and ICAM-1, or TNF-α and sPD-L1) improved predictive accuracy. Univariate analysis identified IL-8 and ICAM-1 as potential markers to predict PFS. Multivariate analysis suggested that high level of IL-8 (P =0.010) and low level of ICAM-1 (P =0.011) correlated significantly with a longer PFS. Conclusion IL-8 and ICAM-1 in plasma have the potential to predict objective response and PFS in patients with locally advanced NSCLC underwent chemoradiotherapy.
Collapse
Affiliation(s)
- Xin Sui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Leilei Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Huajing Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lan Mi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Lymphoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bo Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Anhui Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Rong Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dongming Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Huiming Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Weihu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
8
|
Desideri I, Loi M, Francolini G, Becherini C, Livi L, Bonomo P. Application of Radiomics for the Prediction of Radiation-Induced Toxicity in the IMRT Era: Current State-of-the-Art. Front Oncol 2020; 10:1708. [PMID: 33117669 PMCID: PMC7574641 DOI: 10.3389/fonc.2020.01708] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
Normal tissue complication probability (NTCP) models that were formulated in the Quantitative Analyses of Normal Tissue Effects in the Clinic (QUANTEC) are one of the pillars in support of everyday’s clinical radiation oncology. Because of steady therapeutic refinements and the availability of cutting-edge technical solutions, the ceiling of organs-at-risk-sparing has been reached for photon-based intensity modulated radiotherapy (IMRT). The possibility to capture heterogeneity of patients and tissues in the prediction of toxicity is still an unmet need in modern radiation therapy. Potentially, a major step towards a wider therapeutic index could be obtained from refined assessment of radiation-induced morbidity at an individual level. The rising integration of quantitative imaging and machine learning applications into radiation oncology workflow offers an unprecedented opportunity to further explore the biologic interplay underlying the normal tissue response to radiation. Based on these premises, in this review we focused on the current-state-of-the-art on the use of radiomics for the prediction of toxicity in the field of head and neck, lung, breast and prostate radiotherapy.
Collapse
Affiliation(s)
- Isacco Desideri
- Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Mauro Loi
- Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Giulio Francolini
- Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Carlotta Becherini
- Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Lorenzo Livi
- Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Pierluigi Bonomo
- Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Chargari C, Levy A, Paoletti X, Soria JC, Massard C, Weichselbaum RR, Deutsch E. Methodological Development of Combination Drug and Radiotherapy in Basic and Clinical Research. Clin Cancer Res 2020; 26:4723-4736. [PMID: 32409306 DOI: 10.1158/1078-0432.ccr-19-4155] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 05/12/2020] [Indexed: 01/03/2023]
Abstract
Newer technical improvements in radiation oncology have been rapidly implemented in recent decades, allowing an improved therapeutic ratio. The development of strategies using local and systemic treatments concurrently, mainly targeted therapies, has however plateaued. Targeted molecular compounds and immunotherapy are increasingly being incorporated as the new standard of care for a wide array of cancers. A better understanding of possible prior methodology issues is therefore required and should be integrated into upcoming early clinical trials including individualized radiotherapy-drug combinations. The outcome of clinical trials is influenced by the validity of the preclinical proofs of concept, the impact on normal tissue, the robustness of biomarkers and the quality of the delivery of radiation. Herein, key methodological aspects are discussed with the aim of optimizing the design and implementation of future precision drug-radiotherapy trials.
Collapse
Affiliation(s)
- Cyrus Chargari
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Xavier Paoletti
- University of Versailles St. Quentin, France
- Institut Curie INSERM U900, Biostatistics for Personalized Medicine Team, St. Cloud, France
| | | | - Christophe Massard
- Université Paris-Sud, Orsay, France
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
10
|
Oliver DE, Mohammadi H, Figura N, Frakes JM, Yamoah K, Perez BA, Wuthrick EJ, Naghavi AO, Caudell JJ, Harrison LB, Torres-Roca JF, Ahmed KA. Novel Genomic-Based Strategies to Personalize Lymph Node Radiation Therapy. Semin Radiat Oncol 2019; 29:111-125. [DOI: 10.1016/j.semradonc.2018.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
11
|
Kong FMS, Hirsch FR, Machtay M. Potential future consideration for imaging and blood-based biomarkers for precision medicine in lung cancer. Transl Lung Cancer Res 2017; 6:713-715. [PMID: 29218273 DOI: 10.21037/tlcr.2017.09.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Feng-Ming Spring Kong
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IU, USA
| | - Fred R Hirsch
- Division of Medical Oncology, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Mitchell Machtay
- Department of Radiation Oncology, Case Western University Hospital, Cleveland, OH, USA
| |
Collapse
|