1
|
Cafiero C, Palmirotta R, Martinelli C, Micera A, Giacò L, Persiani F, Morrione A, Pastore C, Nisi C, Modoni G, Galeano T, Guarino T, Foggetti I, Nisticò C, Giordano A, Pisconti S. Oncological Treatment Adverse Reaction Prediction: Development and Initial Validation of a Pharmacogenetic Model in Non-Small-Cell Lung Cancer Patients. Genes (Basel) 2025; 16:265. [PMID: 40149417 PMCID: PMC11942520 DOI: 10.3390/genes16030265] [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: 01/09/2025] [Revised: 02/13/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: The accurate prediction of adverse drug reactions (ADRs) to oncological treatments still poses a clinical challenge. Chemotherapy is usually selected based on clinical trials that do not consider patient variability in ADR risk. Consequently, many patients undergo multiple treatments to find the appropriate medication or dosage, enhancing ADR risks and increasing the chance of discontinuing therapy. We first aimed to develop a pharmacogenetic model for predicting chemotherapy-induced ADRs in cancer patients (the ANTIBLASTIC DRUG MULTIPANEL PLATFORM) and then to assess its feasibility and validate this model in patients with non-small-cell lung cancer (NSCLC) undergoing oncological treatments. Methods: Seventy NSCLC patients of all stages that needed oncological treatment at our facility were enrolled, reflecting the typical population served by our institution, based on geographic and demographic characteristics. Treatments followed existing guidelines, and patients were continuously monitored for adverse reactions. We developed and used a multipanel platform based on 326 SNPs that we identified as strongly associated with response to cancer treatments. Subsequently, a network-based algorithm to link these SNPs to molecular and biological functions, as well as efficacy and adverse reactions to oncological treatments, was used. Results: Data and blood samples were collected from 70 NSCLC patients. A bioinformatic analysis of all identified SNPs highlighted five clusters of patients based on variant aggregations and the associated genes, suggesting potential susceptibility to treatment-related toxicity. We assessed the feasibility of the platform and technically validated it by comparing NSCLC patients undergoing the same course of treatment with or without ADRs against the cluster combination. An odds ratio analysis confirmed the correlation between cluster allocation and increased ADR risk, indicating specific treatment susceptibilities. Conclusions: The ANTIBLASTIC DRUG MULTIPANEL PLATFORM was easily applicable and able to predict ADRs in NSCLC patients undergoing oncological treatments. The application of this novel predictive model could significantly reduce adverse drug reactions and improve the rate of chemotherapy completion, enhancing patient outcomes and quality of life. Its potential for broader prescription management suggests significant treatment improvements in cancer patients.
Collapse
Affiliation(s)
- Concetta Cafiero
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
- Anatomic Pathology Unit, Fabrizio Spaziani Hospital, 03100 Frosinone, Italy
| | - Raffaele Palmirotta
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Canio Martinelli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Department of Biology College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (C.M.); (A.M.); (A.G.)
- Gynecology and Obstetrics Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, 00184 Rome, Italy;
| | - Luciano Giacò
- Bioinformatics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (F.P.)
| | - Federica Persiani
- Bioinformatics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (F.P.)
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Department of Biology College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (C.M.); (A.M.); (A.G.)
| | - Cosimo Pastore
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| | - Claudia Nisi
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| | - Gabriella Modoni
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| | - Teresa Galeano
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| | - Tiziana Guarino
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| | - Ilaria Foggetti
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| | - Cecilia Nisticò
- Medical Oncology Unit, ASL Frosinone, 03100 Frosinone, Italy;
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Department of Biology College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (C.M.); (A.M.); (A.G.)
- Department of Medical Biotechnology, University of Siena, 53100 Siena, Italy
| | - Salvatore Pisconti
- Medical Oncology, SG Moscati Hospital, 74010 Statte, Italy; (C.C.); (C.P.); (C.N.); (G.M.); (T.G.); (T.G.); (I.F.); (S.P.)
| |
Collapse
|
2
|
Tao Q, Li X, Xia Y, Zheng B, Yan Y, Wang S, Jia L. LINC00261 triggers DNA damage via the miR-23a-3p/CELF2 axis to mitigate the malignant characteristics of 131I-resistant papillary thyroid carcinoma cells. Biochem Biophys Rep 2024; 40:101858. [PMID: 39552712 PMCID: PMC11564912 DOI: 10.1016/j.bbrep.2024.101858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/11/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024] Open
Abstract
Background Long-chain non-coding RNA (LINC00261) in the treatment of papillary thyroid carcinoma (PTC) with 131I is still unknown despite its proven anti-tumour effect in thyroid cancer (TC) and other types of cancer. Methods The database and RT-qPCR were used to analyze the expression level of LINC00261 in PTC and cell lines. PTC cells resistant to 131I (TPC-1/R) were created through ongoing exposure to a lethal dose of 131I, and a subcutaneous xenotransplantation model was developed using PTC mice. Bioinformatics analysis and dual-luciferase assays demonstrated the interaction between LINC00261, miR-23a-3p, and CELF2. RT-qPCR and Western blot were used to detect the expression of LINC00261, miR-23a-3p, and CELF2. Additionally, CCK-8, flow cytometry, immunofluorescence (IF), Western blot, and comet assay were employed to measure cell viability level and DNA damage. Results PTC cell lines exhibited a decrease in the expression of LINC00261. The growth and progression through the S-phase of TPC-1/R cells were suppressed by LINC00261, leading to increased apoptosis and DNA damage. The objective of LINC00261 was to regulate the axis of miR-23a-3p/CELF2. Downregulating LINC00261 enhances the growth and advancement of 131I-resistant cells in the S-phase by activating the miR-23a-3p/CELF2 pathway while suppressing cell death and DNA harm. The miR-23a-3p/CELF2 axis activates DNA damage in 131I-resistant PTC cells by LINC00261. Conclusions LINC00261 activates DNA damage in 131I-resistant PTC cells caused by miR-23a-3p/CELF2 axis, improving the progression of cancer cells of PTC.
Collapse
Affiliation(s)
- Qingyuan Tao
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Xiaojin Li
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Central Laboratory, Kunming, Yunnan, 650021, China
| | - Yanyan Xia
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Bin Zheng
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Yijun Yan
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Songrun Wang
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Li Jia
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| |
Collapse
|
3
|
Ren L, Chen DB, Yan X, She S, Yang Y, Zhang X, Liao W, Chen H. Bridging the Gap Between Imaging and Molecular Characterization: Current Understanding of Radiomics and Radiogenomics in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:2359-2372. [PMID: 39619602 PMCID: PMC11608547 DOI: 10.2147/jhc.s423549] [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: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide and the third leading cause of cancer-related deaths. Imaging plays a crucial role in the screening, diagnosis, and monitoring of HCC; however, the potential mechanism regarding phenotypes or molecular subtyping remains underexplored. Radiomics significantly expands the selection of features available by extracting quantitative features from imaging data. Radiogenomics bridges the gap between imaging and genetic/transcriptomic information by associating imaging features with critical genes and pathways, thereby providing biological annotations to these features. Despite challenges in interpreting these connections, assessing their universality, and considering the diversity in HCC etiology and genetic information across different populations, radiomics and radiogenomics offer new perspectives for precision treatment in HCC. This article provides an up-to-date summary of the advancements in radiomics and radiogenomics throughout the HCC care continuum, focusing on the clinical applications, advantages, and limitations of current techniques and offering prospects. Future research should aim to overcome these challenges to improve the prognosis of HCC patients and leverage imaging information for patient benefit.
Collapse
Affiliation(s)
- Liying Ren
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Dong Bo Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Xuanzhi Yan
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People’s Republic of China
| | - Shaoping She
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Yao Yang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Xue Zhang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People’s Republic of China
| | - Hongsong Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| |
Collapse
|
4
|
Aguiar BRL, Ferreira EB, Normando AGC, Dias SDS, Guerra ENS, Reis PED. Potential Single Nucleotide Polymorphisms markers for radiation dermatitis in head and neck cancer patients: a meta-analysis. Strahlenther Onkol 2024; 200:568-582. [PMID: 38668865 DOI: 10.1007/s00066-024-02237-3] [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: 01/08/2024] [Accepted: 04/07/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE To identify potential Single Nucleotide Polymorphisms (SNPs) of susceptibility for the development of acute radiation dermatitis in head and neck cancer patients, and also to verify the association between SNPs and the severity of RD. METHODS This systematic review was reported according to the PRISMA guideline. The proportion meta-analysis was performed to identify the prevalence of genetic markers by geographical region and radiation dermatitis severity. The meta-analysis was performed to verify the association between genetic markers and RD severity. The certainty of the evidence was assessed by GRADE. RESULTS Thirteen studies were included. The most prevalent SNPs were XRCC3 (rs861639) (36%), TGFβ1 (rs1800469) (35%), and RAD51 (rs1801321) (34%). There are prevalence studies in Europe and Asia, with a similar prevalence for all SNPs (29-40%). The prevalence was higher in patients who developed radiation dermatitis ≤2 for any subtype of genes (75-76%). No SNP showed a statistically significant association with very low certainty of evidence. CONCLUSION The most prevalent SNPs may be predictors of acute RD. The analysis of SNP before starting radiation therapy may be a promising method to predict the risk of developing radiation dermatitis and allow radiosensitive patients to have a customized treatment. This current review provides new research directions.
Collapse
Affiliation(s)
- Beatriz Regina Lima Aguiar
- Health Science Graduate Program, School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Elaine Barros Ferreira
- Health Science Graduate Program, School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
- Nursing Department, School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | | | | | - Eliete Neves Silva Guerra
- Laboratory of Oral Histopathology, School of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Paula Elaine Diniz Reis
- Nursing Department, School of Health Sciences, University of Brasilia, Brasília, DF, Brazil.
- School of Health Sciences, Campus Darcy Ribeiro, Asa Norte, University of Brasilia, 70910-900, Brasília, DF, Brazil.
| |
Collapse
|
5
|
Di Pilla A, Nero C, Specchia ML, Ciccarone F, Boldrini L, Lenkowicz J, Alberghetti B, Fagotti A, Testa AC, Valentini V, Sala E, Scambia G. A cost-effectiveness analysis of an integrated clinical-radiogenomic screening program for the identification of BRCA 1/2 carriers (e-PROBE study). Sci Rep 2024; 14:928. [PMID: 38195911 PMCID: PMC10776619 DOI: 10.1038/s41598-023-51031-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 12/29/2023] [Indexed: 01/11/2024] Open
Abstract
Current approach to identify BRCA 1/2 carriers in the general population is ineffective as most of the carriers remain undiagnosed. Radiomics is an emerging tool for large scale quantitative analysis of features from standard diagnostic imaging and has been applied also to identify gene mutational status. The objective of this study was to evaluate the clinical and economic impact of integrating a radiogenomics model with clinical and family history data in identifying BRCA mutation carriers in the general population. This cost-effective analysis compares three different approaches to women selection for BRCA testing: established clinical criteria/family history (model 1); established clinical criteria/family history and the currently available radiogenomic model (49% sensitivity and 87% specificity) based on ultrasound images (model 2); same approach used in model 2 but simulating an improvement of the performances of the radiogenomic model (80% sensitivity and 95% specificity) (model 3). All models were trained with literature data. Direct costs were calculated according to the rates currently used in Italy. The analysis was performed simulating different scenarios on the generation of 18-year-old girls in Italy (274,000 people). The main outcome was to identify the most effective model comparing the number of years of BRCA-cancer healthy life expectancy (HLYs). An incremental cost-effectiveness ratio (ICER) was also derived to determine the cost in order to increase BRCA carriers-healthy life span by 1 year. Compared to model 1, model 2 increases the detection rate of BRCA carriers by 41.8%, reduces the rate of BRCA-related cancers by 23.7%, generating over a 62-year observation period a cost increase by 2.51 €/Year/Person. Moreover, model 3 further increases BRCA carriers detection (+ 68.3%) and decrease in BRCA-related cancers (- 38.4%) is observed compared to model 1. Model 3 increases costs by 0.7 €/Year/Person. After one generation, the estimated ICER in the general population amounts to about 3800€ and 653€ in model 2 and model 3 respectively. Model 2 has a massive effect after only one generation in detecting carriers in the general population with only a small cost increment. The clinical impact is limited mainly due to the current low acceptance rate of risk-reducing surgeries. Further multicentric studies are required before implementing the integrated clinical-radiogenomic model in clinical practice.
Collapse
Affiliation(s)
- A Di Pilla
- Dipartimento di Scienze della Vita e Sanità Pubblica - Sezione di Igiene, Università Cattolica del Sacro Cuore, Rome, Italy
| | - C Nero
- UOC Ginecologia Oncologica, Dipartimento per le Scienze della salute della donna, del Bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - M L Specchia
- Dipartimento di Scienze della Vita e Sanità Pubblica - Sezione di Igiene, Università Cattolica del Sacro Cuore, Rome, Italy.
- Università Cattolica del Sacro Cuore, Rome, Italy.
| | - F Ciccarone
- UOC Ginecologia Oncologica, Dipartimento per le Scienze della salute della donna, del Bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - L Boldrini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Radiomics Research Core Facility, Gemelli Science and Technology Park, Rome, Italy
| | - J Lenkowicz
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Radiomics Research Core Facility, Gemelli Science and Technology Park, Rome, Italy
| | - B Alberghetti
- UOC Ginecologia Oncologica, Dipartimento per le Scienze della salute della donna, del Bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - A Fagotti
- UOC Ginecologia Oncologica, Dipartimento per le Scienze della salute della donna, del Bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - A C Testa
- UOC Ginecologia Oncologica, Dipartimento per le Scienze della salute della donna, del Bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - V Valentini
- Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Radiomics Research Core Facility, Gemelli Science and Technology Park, Rome, Italy
| | - E Sala
- Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Radiologia, Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - G Scambia
- UOC Ginecologia Oncologica, Dipartimento per le Scienze della salute della donna, del Bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
6
|
Jelonek K, Mrowiec K, Gabryś D, Widłak P. The Metabolic Footprint of Systemic Effects in the Blood Caused by Radiotherapy and Inflammatory Conditions: A Systematic Review. Metabolites 2023; 13:1000. [PMID: 37755280 PMCID: PMC10534379 DOI: 10.3390/metabo13091000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/27/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Response to radiotherapy (RT) includes tissue toxicity, which may involve inflammatory reactions. We aimed to compare changes in metabolic patterns induced at the systemic level by radiation and inflammation itself. Patients treated with RT due to head and neck cancer and patients with inflammation-related diseases located in the corresponding anatomical regions were selected. PubMed and Web of Science databases were searched from 1 January 2000 to 10 August 2023. Twenty-five relevant studies where serum/plasma metabolic profiles were analyzed using different metabolomics approaches were identified. The studies showed different metabolic patterns of acute and chronic inflammatory diseases, yet changes in metabolites linked to the urea cycle and metabolism of arginine and proline were common features of both conditions. Although the reviewed reports showed only a few specific metabolites common for early RT response and inflammatory diseases, partly due to differences in metabolomics approaches, several common metabolic pathways linked to metabolites affected by radiation and inflammation were revealed. They included pathways involved in energy metabolism (e.g., metabolism of ketone bodies, mitochondrial electron transport chain, Warburg effect, citric acid cycle, urea cycle) and metabolism of certain amino acids (Arg, Pro, Gly, Ser, Met, Ala, Glu) and lipids (glycerolipids, branched-chain fatty acids). However, metabolites common for RT and inflammation-related diseases could show opposite patterns of changes. This could be exemplified by the lysophosphatidylcholine to phosphatidylcholine ratio (LPC/PC) that increased during chronic inflammation and decreased during the early phase of response to RT. One should be aware of dynamic metabolic changes during different phases of response to radiation, which involve increased levels of LPC in later phases. Hence, metabolomics studies that would address molecular features of both types of biological responses using comparable analytical and clinical approaches are needed to unravel the complexities of these phenomena, ultimately contributing to a deeper understanding of their impact on biological systems.
Collapse
Affiliation(s)
- Karol Jelonek
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-100 Gliwice, Poland;
| | - Katarzyna Mrowiec
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-100 Gliwice, Poland;
| | - Dorota Gabryś
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-100 Gliwice, Poland;
| | - Piotr Widłak
- 2nd Department of Radiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| |
Collapse
|
7
|
Ren Y, Bo L, Shen B, Yang J, Xu S, Shen W, Chen H, Wang X, Chen H, Cai X. Development and validation of a clinical-radiomics model to predict recurrence for patients with hepatocellular carcinoma after curative resection. Med Phys 2023; 50:778-790. [PMID: 36269204 DOI: 10.1002/mp.16061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Recurrence is the leading cause of death in hepatocellular carcinoma (HCC) patients with curative resection. In this study, we aimed to develop a preoperative predictive model based on high-throughput radiomics features and clinical factors for prediction of long- and short-term recurrence for these patients. METHODS A total of 270 patients with HCC who were followed up for at least 5 years after curative hepatectomy between June 2014 and December 2017 were enrolled in this retrospective study. Regions of interest were manually delineated in preoperative T2-weighted images using ITK-SNAP software on each HCC tumor slice. A total of 1197 radiomics features were extracted, and the recursive feature elimination method based on logistic regression was used for radiomics signature building. Tenfold cross-validation was applied for model development. Nomograms were constructed and assessed by calibration plot, which compares nomogram-predicated probability with observed outcome. Receiver-operating characteristic was then generated to evaluate the predictive performance of the model in the development and test cohorts. RESULTS The 10 most recurrence-free survival-related radiomics features were selected for the radiomics signatures. A multiparametric clinical-radiomics model combining albumin and radiomics score for recurrence prediction was further established. The integrated model demonstrated good calibration and satisfactory discrimination, with the area under the curve (AUC) of 0.864, 95% CI 0.842-0.903, sensitivity of 0.889, and specificity of 0.644 in the test set. Calibration curve showed good agreement concerning 5-year recurrence risk predicted by the nomogram. In addition, the AUC of 1-, 2-, 3-, and 4-year recurrence was 0.935 (95% CI 0.836-1.000), 0.861 (95% CI 0.723-0.999), 0.878 (95% CI 0.762-0.994), and 0.878 (95% CI 0.762-0.994) in the test set, respectively. CONCLUSIONS The clinical-radiomics model integrating radiomics features and clinical factors can improve recurrence predictions beyond predictions made using clinical factors or radiomics features alone. Our clinical-radiomics model is a valid method to predict recurrence that should improve preoperative prognostic performance and allow more individualized treatment decisions.
Collapse
Affiliation(s)
- Yiyue Ren
- Department of General Surgery, Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linlin Bo
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, Shandong, China
| | - Bo Shen
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Radiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, China
| | - Jing Yang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shufeng Xu
- Department of Radiology, People's Hospital of Quzhou, Quzhou Hospital Affiliated to Wenzhou Medical University, Quzhou, Zhejiang, China
| | - Weiqiang Shen
- Department of Radiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, China
| | - Hao Chen
- Department of Radiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, China
| | - Xiaoyan Wang
- Department of Medical Imaging, Bengbu Medical College, Bengbu, Anhui, China
| | - Haipeng Chen
- Deepwise Artificial Intelligence Laboratory, Beijing, China
| | - Xiujun Cai
- Department of General Surgery, Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Sminia P, Guipaud O, Viktorsson K, Ahire V, Baatout S, Boterberg T, Cizkova J, Dostál M, Fernandez-Palomo C, Filipova A, François A, Geiger M, Hunter A, Jassim H, Edin NFJ, Jordan K, Koniarová I, Selvaraj VK, Meade AD, Milliat F, Montoro A, Politis C, Savu D, Sémont A, Tichy A, Válek V, Vogin G. Clinical Radiobiology for Radiation Oncology. RADIOBIOLOGY TEXTBOOK 2023:237-309. [DOI: 10.1007/978-3-031-18810-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
AbstractThis chapter is focused on radiobiological aspects at the molecular, cellular, and tissue level which are relevant for the clinical use of ionizing radiation (IR) in cancer therapy. For radiation oncology, it is critical to find a balance, i.e., the therapeutic window, between the probability of tumor control and the probability of side effects caused by radiation injury to the healthy tissues and organs. An overview is given about modern precision radiotherapy (RT) techniques, which allow optimal sparing of healthy tissues. Biological factors determining the width of the therapeutic window are explained. The role of the six typical radiobiological phenomena determining the response of both malignant and normal tissues in the clinic, the 6R’s, which are Reoxygenation, Redistribution, Repopulation, Repair, Radiosensitivity, and Reactivation of the immune system, is discussed. Information is provided on tumor characteristics, for example, tumor type, growth kinetics, hypoxia, aberrant molecular signaling pathways, cancer stem cells and their impact on the response to RT. The role of the tumor microenvironment and microbiota is described and the effects of radiation on the immune system including the abscopal effect phenomenon are outlined. A summary is given on tumor diagnosis, response prediction via biomarkers, genetics, and radiomics, and ways to selectively enhance the RT response in tumors. Furthermore, we describe acute and late normal tissue reactions following exposure to radiation: cellular aspects, tissue kinetics, latency periods, permanent or transient injury, and histopathology. Details are also given on the differential effect on tumor and late responding healthy tissues following fractionated and low dose rate irradiation as well as the effect of whole-body exposure.
Collapse
|
9
|
Helm A, Totis C, Durante M, Fournier C. Are charged particles a good match for combination with immunotherapy? Current knowledge and perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:1-36. [PMID: 36997266 DOI: 10.1016/bs.ircmb.2023.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Charged particle radiotherapy, mainly using protons and carbon ions, provides physical characteristics allowing for a volume conformal irradiation and a reduction of the integral dose to normal tissue. Carbon ion therapy additionally features an increased biological effectiveness resulting in peculiar molecular effects. Immunotherapy, mostly performed with immune checkpoint inhibitors, is nowadays considered a pillar in cancer therapy. Based on the advantageous features of charged particle radiotherapy, we review pre-clinical evidence revealing a strong potential of its combination with immunotherapy. We argue that the combination therapy deserves further investigation with the aim of translation in clinics, where a few studies have been set up already.
Collapse
Affiliation(s)
- A Helm
- Biophysics Department, GSI, Darmstadt, Germany
| | - C Totis
- Biophysics Department, GSI, Darmstadt, Germany
| | - M Durante
- Biophysics Department, GSI, Darmstadt, Germany.
| | - C Fournier
- Biophysics Department, GSI, Darmstadt, Germany
| |
Collapse
|
10
|
Hu Q, Li K, Yang C, Wang Y, Huang R, Gu M, Xiao Y, Huang Y, Chen L. The role of artificial intelligence based on PET/CT radiomics in NSCLC: Disease management, opportunities, and challenges. Front Oncol 2023; 13:1133164. [PMID: 36959810 PMCID: PMC10028142 DOI: 10.3389/fonc.2023.1133164] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Objectives Lung cancer has been widely characterized through radiomics and artificial intelligence (AI). This review aims to summarize the published studies of AI based on positron emission tomography/computed tomography (PET/CT) radiomics in non-small-cell lung cancer (NSCLC). Materials and methods A comprehensive search of literature published between 2012 and 2022 was conducted on the PubMed database. There were no language or publication status restrictions on the search. About 127 articles in the search results were screened and gradually excluded according to the exclusion criteria. Finally, this review included 39 articles for analysis. Results Classification is conducted according to purposes and several studies were identified at each stage of disease:1) Cancer detection (n=8), 2) histology and stage of cancer (n=11), 3) metastases (n=6), 4) genotype (n=6), 5) treatment outcome and survival (n=8). There is a wide range of heterogeneity among studies due to differences in patient sources, evaluation criteria and workflow of radiomics. On the whole, most models show diagnostic performance comparable to or even better than experts, and the common problems are repeatability and clinical transformability. Conclusion AI-based PET/CT Radiomics play potential roles in NSCLC clinical management. However, there is still a long way to go before being translated into clinical application. Large-scale, multi-center, prospective research is the direction of future efforts, while we need to face the risk of repeatability of radiomics features and the limitation of access to large databases.
Collapse
Affiliation(s)
- Qiuyuan Hu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Conghui Yang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yue Wang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Rong Huang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Mingqiu Gu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yuqiang Xiao
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| | - Long Chen
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| |
Collapse
|
11
|
miR-193a-5p Enhances the Radioresistance of Pancreatic Cancer Cells by Targeting ZFP57 and Activating the Wnt Pathway. JOURNAL OF ONCOLOGY 2022; 2022:8071343. [PMID: 36276285 PMCID: PMC9586754 DOI: 10.1155/2022/8071343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
Abstract
This study was to investigate whether miR-193a-5p and ZFP57 are involved in the radioresistance of pancreatic cancer and to explore its working mechanism. Pancreatic cancer tissues were harvested from patients who achieved CR (complete remission) and PR (partial remission) and those who achieved PD (progressive disease) and SD (stable disease). The mRNA and protein expressions of ZFP57 and miR-193a-5p were determined by RT-qPCR and WB (Western blot), respectively. For in vitro experiments, the parental BxPC-3 cell line was irradiated by X-ray at a total dose of 40 Gy to induce the irradiation-resistant subtype BxPC-3-RR. ZFP57 was downregulated in radioresistant pancreatic cancer cells. The results of dual-luciferase reporter gene assay, RNA pull-down assay, RT-qPCR, and WB confirmed that miR-193a-5p targeted ZFP57 and inhibited ZFP57 expression. The MTT assay and the colony formation assay showed that the radioresistant pancreatic cancer cells had higher viability and survival fraction. The results of WB indicated that in the radioresistant pancreatic cancer cells, the cyclin D1, Bax, CDk4, cleaved caspase-3, Bcl-2, and γ-H2AX proteins were upregulated to varying degrees. The results of the in vitro nude mouse experiment were consistent with those of in vivo experiments. According to the cell transfection and salvage experiments, miR-193a-5p down regulated ZFP57 after radiotherapy. As a result, the Wnt pathway was activated, which further induced radioresistance of pancreatic cancer cells. Our experiments showed that the miR-193a-5p/ZFP57/Wnt pathway mediated the radioresistance of pancreatic cancer cells, providing novel clues for the treatment of pancreatic cancer.
Collapse
|
12
|
Li G, Wu X, Ma X. Artificial intelligence in radiotherapy. Semin Cancer Biol 2022; 86:160-171. [PMID: 35998809 DOI: 10.1016/j.semcancer.2022.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/18/2022] [Indexed: 11/19/2022]
Abstract
Radiotherapy is a discipline closely integrated with computer science. Artificial intelligence (AI) has developed rapidly over the past few years. With the explosive growth of medical big data, AI promises to revolutionize the field of radiotherapy through highly automated workflow, enhanced quality assurance, improved regional balances of expert experiences, and individualized treatment guided by multi-omics. In addition to independent researchers, the increasing number of large databases, biobanks, and open challenges significantly facilitated AI studies on radiation oncology. This article reviews the latest research, clinical applications, and challenges of AI in each part of radiotherapy including image processing, contouring, planning, quality assurance, motion management, and outcome prediction. By summarizing cutting-edge findings and challenges, we aim to inspire researchers to explore more future possibilities and accelerate the arrival of AI radiotherapy.
Collapse
Affiliation(s)
- Guangqi Li
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xin Wu
- Head & Neck Oncology ward, Division of Radiotherapy Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China.
| |
Collapse
|
13
|
Khaleel S, Katims A, Cumarasamy S, Rosenzweig S, Attalla K, Hakimi AA, Mehrazin R. Radiogenomics in Clear Cell Renal Cell Carcinoma: A Review of the Current Status and Future Directions. Cancers (Basel) 2022; 14:2085. [PMID: 35565216 PMCID: PMC9100795 DOI: 10.3390/cancers14092085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/30/2022] Open
Abstract
Radiogenomics is a field of translational radiology that aims to associate a disease's radiologic phenotype with its underlying genotype, thus offering a novel class of non-invasive biomarkers with diagnostic, prognostic, and therapeutic potential. We herein review current radiogenomics literature in clear cell renal cell carcinoma (ccRCC), the most common renal malignancy. A literature review was performed by querying PubMed, Medline, Cochrane Library, Google Scholar, and Web of Science databases, identifying all relevant articles using the following search terms: "radiogenomics", "renal cell carcinoma", and "clear cell renal cell carcinoma". Articles included were limited to the English language and published between 2009-2021. Of 141 retrieved articles, 16 fit our inclusion criteria. Most studies used computed tomography (CT) images from open-source and institutional databases to extract radiomic features that were then modeled against common genomic mutations in ccRCC using a variety of machine learning algorithms. In more recent studies, we noted a shift towards the prediction of transcriptomic and/or epigenetic disease profiles, as well as downstream clinical outcomes. Radiogenomics offers a platform for the development of non-invasive biomarkers for ccRCC, with promising results in small-scale retrospective studies. However, more research is needed to identify and validate robust radiogenomic biomarkers before integration into clinical practice.
Collapse
Affiliation(s)
- Sari Khaleel
- Memorial Sloan Kettering Cancer Center, Department of Urology, New York, NY 10065, USA; (S.K.); (A.A.H.)
| | - Andrew Katims
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.K.); (S.C.); (S.R.); (K.A.)
| | - Shivaram Cumarasamy
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.K.); (S.C.); (S.R.); (K.A.)
| | - Shoshana Rosenzweig
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.K.); (S.C.); (S.R.); (K.A.)
| | - Kyrollis Attalla
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.K.); (S.C.); (S.R.); (K.A.)
| | - A Ari Hakimi
- Memorial Sloan Kettering Cancer Center, Department of Urology, New York, NY 10065, USA; (S.K.); (A.A.H.)
| | - Reza Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.K.); (S.C.); (S.R.); (K.A.)
| |
Collapse
|
14
|
Kambouris ME, Goudoudaki S, Kritikou S, Milioni A, Karamperis K, Giavasis I, Patrinos GP, Velegraki A, Manoussopoulos Y. Beyond the Microbiome: Germ-ganism? An Integrative Idea for Microbial Existence, Organization, Growth, Pathogenicity, and Therapeutics. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:204-217. [PMID: 35255221 DOI: 10.1089/omi.2022.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The advances made by microbiome research call for new vocabulary and expansion of our thinking in microbiology. For example, the life-forms presenting in both unicellular and multicellular formats invite us to rethink microbial existence, organization, growth, pathogenicity, and therapeutics in the 21st century. A view of such populations as parts of single organisms with a loose, distributed multicellular organization, introduced here as a germ-ganism, rather than communities, might open up interesting prospects for diagnostics and therapeutics innovation. This study tested and further contextualized the concept of germ-ganism using solid cultures of bacteria and fungi. Based on our findings and the literature reviewed herein, we propose that germ-ganism has synergy with a systems medicine approach by broadening host-environment interactions from cells and microorganisms to a scale of biological ecosystems. Germ-ganism also brings about the possibility of studying the multilevel impacts of novel therapeutic agents within and across networks of microbial ecosystems. The germ-ganism would lend itself, in the long term, to a veritable biocybernetics system, while in the mid-term, we anticipate it will contribute to new diagnostics and therapeutics. Biosecurity applications would be immensely affected by germ-ganism. Industrial applications of germ-ganism are of interest as a more sustainable alternative to costly solutions such as tampered strains/microorganisms. In conclusion, germ-ganism is informed by lessons from microbiome research and invites rethinking microbial existence, organization, and growth as an organism. Germ-ganism has vast ramifications for understanding pathogenicity, and clinical, biosecurity, and biotechnology applications in the current historical moment of the COVID-19 pandemic and beyond.
Collapse
Affiliation(s)
- Manousos E Kambouris
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | - Stavroula Goudoudaki
- Plant Protection Division of Patras, Institute of Industrial and Forage Plants, Patras, Greece
| | - Stavroula Kritikou
- NCPF/UoA, Laboratory of Microbiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Aphroditi Milioni
- NCPF/UoA, Laboratory of Microbiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Kariofyllis Karamperis
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | - Ioannis Giavasis
- Laboratory of Food Microbiology and Biotechnology, Department of Food Science and Nutrition, University of Thessaly, Karditsa, Greece
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | - Aristea Velegraki
- NCPF/UoA, Laboratory of Microbiology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Yiannis Manoussopoulos
- Plant Protection Division of Patras, Institute of Industrial and Forage Plants, Patras, Greece
| |
Collapse
|
15
|
Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers (Basel) 2022; 14:cancers14071657. [PMID: 35406429 PMCID: PMC8997041 DOI: 10.3390/cancers14071657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool. Abstract Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.
Collapse
Affiliation(s)
- Athanasios K. Anagnostopoulos
- Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
- Correspondence:
| | - Anastasios Gaitanis
- Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Ioannis Gkiozos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Emmanouil I. Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Sofia N. Chatziioannou
- Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Dimitris Thanos
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Achilles N. Chatziioannou
- First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal;
- Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
- The Institute of Cancer Research, London SM2 5MG, UK
- Karolinska Institutet, 14186 Stockholm, Sweden
- Institute of Computer Science, FORTH, 70013 Heraklion, Greece
| |
Collapse
|
16
|
Liu ET, Zhou S, Li Y, Zhang S, Ma Z, Guo J, Guo L, Zhang Y, Guo Q, Xu L. Development and validation of an MRI-based nomogram for the preoperative prediction of tumor mutational burden in lower-grade gliomas. Quant Imaging Med Surg 2022; 12:1684-1697. [PMID: 35284257 PMCID: PMC8899970 DOI: 10.21037/qims-21-300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/30/2021] [Indexed: 09/25/2023]
Abstract
BACKGROUND High tumor mutational burden (TMB) is an emerging biomarker of sensitivity to immune checkpoint inhibitors. In this study, we aimed to determine the value of magnetic resonance (MR)-based preoperative nomogram in predicting TMB status in lower-grade glioma (LGG) patients. METHODS Overall survival (OS) data were derived from The Cancer Genome Atlas (TCGA) and then analyzed by using the Kaplan-Meier method and time-dependent receiver operating characteristic (tdROC) analysis. The magnetic resonance imaging (MRI) data of 168 subjects obtained from The Cancer Imaging Archive (TCIA) were retrospectively analyzed. The correlation was explored by univariate and multivariate regression analyses. Finally, we performed tenfold cross validation. TMB values were retrieved from the supplementary information of a previously published article. RESULTS The high TMB subtype was associated with the shortest median OS (high vs. low: 50.9 vs. 95.6 months, P<0.05). The tdROC for the high-TMB tumors was 74% (95% CI: 61-86%) for survival at 12 months, and 71% (95% CI: 60-82%) for survival at 24 months. Multivariate logistic regression analysis confirmed that three risk factors [extranodular growth: odds ratio (OR): 8.367, 95% CI: 3.153-22.199, P<0.01; length-width ratio ≥ median: OR: 1.947, 95% CI: 1.025-3.697, P<0.05; frontal lobe: OR: 0.455, 95% CI: 0.229-0.903, P<0.05] were significant independent predictors of high-TMB tumors. The nomogram showed good calibration and discrimination. This model had an area under the curve (AUC) of 0.736 (95% CI: 0.655-0.817). Decision curve analysis (DCA) demonstrated that the nomogram was clinically useful. The average accuracy of the tenfold cross validation was 71.6% for high-TMB tumors. CONCLUSIONS Our results indicated that a distinct OS disadvantage was associated with the high TMB group. In addition, extranodular growth, nonfrontal lobe tumors and length-width ratio ≥ median can be conveniently used to facilitate the prediction of high-TMB tumors.
Collapse
Affiliation(s)
- En-Tao Liu
- WeiLun PET Center, Department of Nuclear Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuqin Zhou
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yingwen Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Siwei Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zelan Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Junbiao Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Lei Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yue Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Quanlai Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Li Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
A Novel Four-Gene Prognostic Signature for Prediction of Survival in Patients with Soft Tissue Sarcoma. Cancers (Basel) 2021; 13:cancers13225837. [PMID: 34830998 PMCID: PMC8616347 DOI: 10.3390/cancers13225837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Soft tissue sarcomas (STS) still lack effective clinical stratification and prognostic models. The aim of this study is to establish a reliable prognostic gene signature in STS. Using 189 STS samples from the TCGA database, a four-gene signature (including DHRS3, JRK, TARDBP and TTC3) and nomograms that can be used to predict the overall survival and relapse free survival of STS patients was developed. The predictive ability for metastasis free survival was externally verified in the GEO cohort. We demonstrated that the novel gene signature provides an attractive platform for risk stratification and prognosis prediction of STS patients, which is of great importance for individualized clinical treatment and long-term management of patients with this rare and severe disease. Abstract Soft tissue sarcomas (STS), a group of rare malignant tumours with high tissue heterogeneity, still lack effective clinical stratification and prognostic models. Therefore, we conducted this study to establish a reliable prognostic gene signature. Using 189 STS patients’ data from The Cancer Genome Atlas database, a four-gene signature including DHRS3, JRK, TARDBP and TTC3 was established. A risk score based on this gene signature was able to divide STS patients into a low-risk and a high-risk group. The latter had significantly worse overall survival (OS) and relapse free survival (RFS), and Cox regression analyses showed that the risk score is an independent prognostic factor. Nomograms containing the four-gene signature have also been established and have been verified through calibration curves. In addition, the predictive ability of this four-gene signature for STS metastasis free survival was verified in an independent cohort (309 STS patients from the Gene Expression Omnibus database). Finally, Gene Set Enrichment Analysis indicated that the four-gene signature may be related to some pathways associated with tumorigenesis, growth, and metastasis. In conclusion, our study establishes a novel four-gene signature and clinically feasible nomograms to predict the OS and RFS. This can help personalized treatment decisions, long-term patient management, and possible future development of targeted therapy.
Collapse
|
18
|
de Mey S, Dufait I, De Ridder M. Radioresistance of Human Cancers: Clinical Implications of Genetic Expression Signatures. Front Oncol 2021; 11:761901. [PMID: 34778082 PMCID: PMC8579106 DOI: 10.3389/fonc.2021.761901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Although radiotherapy is given to more than 50% of cancer patients, little progress has been made in identifying optimal radiotherapy - drug combinations to improve treatment efficacy. Using molecular data from The Cancer Genome Atlas (TCGA), we extracted a total of 1016 cancer patients that received radiotherapy. The patients were diagnosed with head-and-neck (HNSC - 294 patients), cervical (CESC - 166 patients) and breast (BRCA - 549 patients) cancer. We analyzed mRNA expression patterns of 50 hallmark gene sets of the MSigDB collection, which we divided in eight categories based on a shared biological or functional process. Tumor samples were split into upregulated, neutral or downregulated mRNA expression for all gene sets using a gene set analysis (GSEA) pre-ranked analysis and assessed for their clinical relevance. We found a prognostic association between three of the eight gene set categories (Radiobiological, Metabolism and Proliferation) and overall survival in all three cancer types. Furthermore, multiple single associations were revealed in the other categories considered. To the best of our knowledge, our study is the first report suggesting clinical relevance of molecular characterization based on hallmark gene sets to refine radiation strategies.
Collapse
Affiliation(s)
- Sven de Mey
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Inès Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
19
|
Ravichandran A, Clegg J, Adams MN, Hampson M, Fielding A, Bray LJ. 3D Breast Tumor Models for Radiobiology Applications. Cancers (Basel) 2021; 13:5714. [PMID: 34830869 PMCID: PMC8616164 DOI: 10.3390/cancers13225714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/28/2021] [Accepted: 11/07/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is a leading cause of cancer-associated death in women. The clinical management of breast cancers is normally carried out using a combination of chemotherapy, surgery and radiation therapy. The majority of research investigating breast cancer therapy until now has mainly utilized two-dimensional (2D) in vitro cultures or murine models of disease. However, there has been significant uptake of three-dimensional (3D) in vitro models by cancer researchers over the past decade, highlighting a complimentary model for studies of radiotherapy, especially in conjunction with chemotherapy. In this review, we underline the effects of radiation therapy on normal and malignant breast cells and tissues, and explore the emerging opportunities that pre-clinical 3D models offer in improving our understanding of this treatment modality.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Julien Clegg
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Mark N. Adams
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Madison Hampson
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
| | - Andrew Fielding
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Laura J. Bray
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| |
Collapse
|
20
|
Coppola F, Faggioni L, Gabelloni M, De Vietro F, Mendola V, Cattabriga A, Cocozza MA, Vara G, Piccinino A, Lo Monaco S, Pastore LV, Mottola M, Malavasi S, Bevilacqua A, Neri E, Golfieri R. Human, All Too Human? An All-Around Appraisal of the "Artificial Intelligence Revolution" in Medical Imaging. Front Psychol 2021; 12:710982. [PMID: 34650476 PMCID: PMC8505993 DOI: 10.3389/fpsyg.2021.710982] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/02/2021] [Indexed: 12/22/2022] Open
Abstract
Artificial intelligence (AI) has seen dramatic growth over the past decade, evolving from a niche super specialty computer application into a powerful tool which has revolutionized many areas of our professional and daily lives, and the potential of which seems to be still largely untapped. The field of medicine and medical imaging, as one of its various specialties, has gained considerable benefit from AI, including improved diagnostic accuracy and the possibility of predicting individual patient outcomes and options of more personalized treatment. It should be noted that this process can actively support the ongoing development of advanced, highly specific treatment strategies (e.g., target therapies for cancer patients) while enabling faster workflow and more efficient use of healthcare resources. The potential advantages of AI over conventional methods have made it attractive for physicians and other healthcare stakeholders, raising much interest in both the research and the industry communities. However, the fast development of AI has unveiled its potential for disrupting the work of healthcare professionals, spawning concerns among radiologists that, in the future, AI may outperform them, thus damaging their reputations or putting their jobs at risk. Furthermore, this development has raised relevant psychological, ethical, and medico-legal issues which need to be addressed for AI to be considered fully capable of patient management. The aim of this review is to provide a brief, hopefully exhaustive, overview of the state of the art of AI systems regarding medical imaging, with a special focus on how AI and the entire healthcare environment should be prepared to accomplish the goal of a more advanced human-centered world.
Collapse
Affiliation(s)
- Francesca Coppola
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
- SIRM Foundation, Italian Society of Medical and Interventional Radiology, Milan, Italy
| | - Lorenzo Faggioni
- Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Michela Gabelloni
- Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Fabrizio De Vietro
- Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Vincenzo Mendola
- Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Arrigo Cattabriga
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Maria Adriana Cocozza
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Giulio Vara
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Alberto Piccinino
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Silvia Lo Monaco
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Luigi Vincenzo Pastore
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Margherita Mottola
- Department of Computer Science and Engineering, University of Bologna, Bologna, Italy
| | - Silvia Malavasi
- Department of Computer Science and Engineering, University of Bologna, Bologna, Italy
| | - Alessandro Bevilacqua
- Department of Computer Science and Engineering, University of Bologna, Bologna, Italy
| | - Emanuele Neri
- SIRM Foundation, Italian Society of Medical and Interventional Radiology, Milan, Italy
- Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Rita Golfieri
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
21
|
Hoshino I, Yokota H. Radiogenomics of gastroenterological cancer: The dawn of personalized medicine with artificial intelligence-based image analysis. Ann Gastroenterol Surg 2021; 5:427-435. [PMID: 34337291 PMCID: PMC8316732 DOI: 10.1002/ags3.12437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/29/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Radiogenomics is a new field of medical science that integrates two omics, radiomics and genomics, and may bring a major paradigm shift in traditional personalized medicine strategies that require tumor tissue samples. In addition, the acquisition of the data does not require special imaging equipment or special imaging conditions, and it is possible to use image information from computed tomography, magnetic resonance imaging, positron emission tomography-computed tomography in clinical practice, so the versatility and cost-effectiveness of radiogenomics are expected. So far, the field of radiogenomics has developed, especially in the fields of brain tumors and breast cancer, but recently, reports of radiogenomic research on gastroenterological cancer are increasing. This review provides an overview of radiogenomic research methods and summarizes the current radiogenomic research in gastroenterological cancer. In addition, the application of artificial intelligence is considered to be indispensable for the integrated analysis of enormous omics information in the future, and the future direction of this research, including the latest technologies, will be discussed.
Collapse
Affiliation(s)
- Isamu Hoshino
- Division of Gastroenterological SurgeryChiba Cancer CenterChibaJapan
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation OncologyGraduate School of MedicineChiba UniversityChibaJapan
| |
Collapse
|
22
|
Jelonek K, Krzywon A, Papaj K, Polanowski P, Szczepanik K, Skladowski K, Widlak P. Dose-dependence of radiotherapy-induced changes in serum levels of choline-containing phospholipids; the importance of lower doses delivered to large volumes of normal tissues. Strahlenther Onkol 2021; 197:926-934. [PMID: 34185114 PMCID: PMC8458179 DOI: 10.1007/s00066-021-01802-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/31/2021] [Indexed: 10/25/2022]
Abstract
BACKGROUND Conformal radiotherapy is a primary treatment in head and neck cancer, which putative adverse effects depend on relatively low doses of radiation delivered to increased volumes of normal tissues. Systemic effects of such treatment include radiation-induced changes in serum lipid profile, yet dose- and volume-dependence of these changes remain to be established. METHODS Here we analyzed levels of choline-containing phospholipids in serum samples collected consecutively during the radiotherapy used as the only treatment modality. The liquid chromatography-mass spectrometry (LC-MS) approach applied in the study enabled the detection and quantitation of 151 phospholipids, including (lyso)phosphatidylcholines and sphingomyelins. RESULTS No statistically significant differences were found in the pretreatment samples from patients with different locations and stages of cancer. To compensate for potential differences between schemes of radiotherapy, the biologically effective doses were calculated and used in the search of correlations with specific lipid levels. We found that the levels of several phospholipids depended on the maximum dose delivered to the gross tumor volume and total radiation energy absorbed by the patient's body. Increased doses correlated with increased levels of sphingomyelins and reduced levels of phosphatidylcholines. Furthermore, we observed several phospholipids whose serum levels correlated with the degree of acute radiation toxicity. CONCLUSION Noteworthy, serum phospholipid levels were associated mainly with volumes of normal tissues irradiated with relatively low doses (i.e., total accumulated dose 20 Gy), which indicated the importance of such effects on the systemic response of the patient's organism to intensity-modulated radiotherapy (IMRT).
Collapse
Affiliation(s)
- Karol Jelonek
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102, Gliwice, Poland.
| | - Aleksandra Krzywon
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102, Gliwice, Poland
| | - Katarzyna Papaj
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100, Gliwice, Poland
| | - Pawel Polanowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102, Gliwice, Poland
| | - Krzysztof Szczepanik
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102, Gliwice, Poland
| | - Krzysztof Skladowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102, Gliwice, Poland
| | - Piotr Widlak
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102, Gliwice, Poland
| |
Collapse
|
23
|
Flint DB, Bright SJ, McFadden CH, Konishi T, Ohsawa D, Turner B, Lin SH, Grosshans DR, Chiu HS, Sumazin P, Shaitelman SF, Sawakuchi GO. Cell lines of the same anatomic site and histologic type show large variability in intrinsic radiosensitivity and relative biological effectiveness to protons and carbon ions. Med Phys 2021; 48:3243-3261. [PMID: 33837540 PMCID: PMC11919485 DOI: 10.1002/mp.14878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To show that intrinsic radiosensitivity varies greatly for protons and carbon (C) ions in addition to photons, and that DNA repair capacity remains important in governing this variability. METHODS We measured or obtained from the literature clonogenic survival data for a number of human cancer cell lines exposed to photons, protons (9.9 keV/μm), and C-ions (13.3-77.1 keV/μm). We characterized their intrinsic radiosensitivity by the dose for 10% or 50% survival (D10% or D50% ), and quantified the variability at each radiation quality by the coefficient of variation (COV) in D10% and D50% . We also treated cells with DNA repair inhibitors prior to irradiation to assess how DNA repair capacity affects their variability. RESULTS We found no statistically significant differences in the COVs of D10% or D50% between any of the radiation qualities investigated. The same was true regardless of whether the cells were treated with DNA repair inhibitors, or whether they were stratified into histologic subsets. Even within histologic subsets, we found remarkable differences in radiosensitivity for high LET C-ions that were often greater than the variations in RBE, with brain cancer cells varying in D10% (D50% ) up to 100% (131%) for 77.1 keV/μm C-ions, and non-small cell lung cancer and pancreatic cancer cell lines varying up to 55% (76%) and 51% (78%), respectively, for 60.5 keV/μm C-ions. The cell lines with modulated DNA repair capacity had greater variability in intrinsic radiosensitivity across all radiation qualities. CONCLUSIONS Even for cell lines of the same histologic type, there are remarkable variations in intrinsic radiosensitivity, and these variations do not differ significantly between photon, proton or C-ion radiation. The importance of DNA repair capacity in governing the variability in intrinsic radiosensitivity is not significantly diminished for higher LET radiation.
Collapse
Affiliation(s)
- David B Flint
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott J Bright
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Conor H McFadden
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Teruaki Konishi
- Single Cell Radiation Biology Group, Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daisuke Ohsawa
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Broderick Turner
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David R Grosshans
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua-Sheng Chiu
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Simona F Shaitelman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel O Sawakuchi
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
lncRNA CASC2 Enhances 131I Sensitivity in Papillary Thyroid Cancer by Sponging miR-155. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7183629. [PMID: 33134385 PMCID: PMC7591961 DOI: 10.1155/2020/7183629] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/14/2020] [Indexed: 11/18/2022]
Abstract
Long noncoding RNA cancer susceptibility candidate 2 (CASC2) has been reported to play an anticancer role in papillary thyroid cancer (PTC). Radioiodine (131I) is a common option for the treatment of PTC. However, the role and mechanism of CASC2 in 131I sensitivity remain unclear. In this study, 131I-resistant cells were constructed through continuous treatment of 131I. The expression levels of CASC2 and miR-155 were measured by qRT-PCR. The IC50 of 131I was analyzed by cell viability using MTT assay. Flow cytometry was conducted to determine cell apoptosis induced by 131I. The association between CASC2 and miR-155 was evaluated by luciferase assay and RNA immunoprecipitation. A mouse xenograft model was built to explore the effect of CASC2 on the growth of 131I-resistant PTC cells in vivo. Results showed that CASC2 expression was decreased in PTC tissues and cells, and low expression of CASC2 was associated with poor outcome of patients. CASC2 level was reduced in 131I-resistant cells. Knockdown of CASC2 inhibited 131I sensitivity in thyroid cancer cells. Overexpression of CASC2 enhanced 131I sensitivity in constructed resistant PTC cells. CASC2 was a decoy of miR-155, and CASC2-mediated promotion of 131I sensitivity was weakened by decreasing miR-155. Abundance of CASC2 inhibited the growth of 131I-resistant cells in vivo. As a conclusion, CASC2 increases 131I sensitivity in PTC by sponging miR-155, providing a novel target for the treatment of thyroid cancer patients with 131I resistance.
Collapse
|
25
|
Tinganelli W, Durante M. Carbon Ion Radiobiology. Cancers (Basel) 2020; 12:E3022. [PMID: 33080914 PMCID: PMC7603235 DOI: 10.3390/cancers12103022] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy using accelerated charged particles is rapidly growing worldwide. About 85% of the cancer patients receiving particle therapy are irradiated with protons, which have physical advantages compared to X-rays but a similar biological response. In addition to the ballistic advantages, heavy ions present specific radiobiological features that can make them attractive for treating radioresistant, hypoxic tumors. An ideal heavy ion should have lower toxicity in the entrance channel (normal tissue) and be exquisitely effective in the target region (tumor). Carbon ions have been chosen because they represent the best combination in this direction. Normal tissue toxicities and second cancer risk are similar to those observed in conventional radiotherapy. In the target region, they have increased relative biological effectiveness and a reduced oxygen enhancement ratio compared to X-rays. Some radiobiological properties of densely ionizing carbon ions are so distinct from X-rays and protons that they can be considered as a different "drug" in oncology, and may elicit favorable responses such as an increased immune response and reduced angiogenesis and metastatic potential. The radiobiological properties of carbon ions should guide patient selection and treatment protocols to achieve optimal clinical results.
Collapse
Affiliation(s)
- Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforchung, Planckstraße 1, 64291 Darmstadt, Germany;
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforchung, Planckstraße 1, 64291 Darmstadt, Germany;
- Institut für Festkörperphysik, Technische Universität Darmstadt, Hochschulstraße 8, 64289 Darmstadt, Germany
| |
Collapse
|
26
|
Nero C, Ciccarone F, Boldrini L, Lenkowicz J, Paris I, Capoluongo ED, Testa AC, Fagotti A, Valentini V, Scambia G. Germline BRCA 1-2 status prediction through ovarian ultrasound images radiogenomics: a hypothesis generating study (PROBE study). Sci Rep 2020; 10:16511. [PMID: 33020566 PMCID: PMC7536234 DOI: 10.1038/s41598-020-73505-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
Radiogenomics is a specific application of radiomics where imaging features are linked to genomic profiles. We aim to develop a radiogenomics model based on ovarian US images for predicting germline BRCA1/2 gene status in women with healthy ovaries. From January 2013 to December 2017 a total of 255 patients addressed to germline BRCA1/2 testing and pelvic US documenting normal ovaries, were retrospectively included. Feature selection for univariate analysis was carried out via correlation analysis. Multivariable analysis for classification of germline BRCA1/2 status was then carried out via logistic regression, support vector machine, ensemble of decision trees and automated machine learning pipelines. Data were split into a training (75%) and a testing (25%) set. The four strategies obtained a similar performance in terms of accuracy on the testing set (from 0.54 of logistic regression to 0.64 of the auto-machine learning pipeline). Data coming from one of the tested US machine showed generally higher performances, particularly with the auto-machine learning pipeline (testing set specificity 0.87, negative predictive value 0.73, accuracy value 0.72 and 0.79 on training set). The study shows that a radiogenomics model on machine learning techniques is feasible and potentially useful for predicting gBRCA1/2 status in women with healthy ovaries.
Collapse
Affiliation(s)
- Camilla Nero
- Dipartimento per le Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology, Rome, Italy.
- Department of Obstetrics and Gynecology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168, Rome, Italy.
| | - Francesca Ciccarone
- Dipartimento per le Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology, Rome, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per immagini, radioterapia oncologica ed ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Jacopo Lenkowicz
- Dipartimento di Diagnostica per immagini, radioterapia oncologica ed ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ida Paris
- Dipartimento per le Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology, Rome, Italy
| | - Ettore Domenico Capoluongo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University-CEINGE, Advanced Biotechnology, Naples, Italy
| | - Antonia Carla Testa
- Dipartimento per le Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology, Rome, Italy
| | - Anna Fagotti
- Dipartimento per le Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology, Rome, Italy
| | - Vincenzo Valentini
- Dipartimento di Diagnostica per immagini, radioterapia oncologica ed ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Dipartimento per le Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Gynecologic Oncology, Rome, Italy
| |
Collapse
|
27
|
Gkika E, Benndorf M, Oerther B, Mohammad F, Beitinger S, Adebahr S, Carles M, Schimek-Jasch T, Zamboglou C, Frye BC, Bamberg F, Waller CF, Werner M, Grosu AL, Nestle U, Kayser G. Immunohistochemistry and Radiomic Features for Survival Prediction in Small Cell Lung Cancer. Front Oncol 2020; 10:1161. [PMID: 32903606 PMCID: PMC7438800 DOI: 10.3389/fonc.2020.01161] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/08/2020] [Indexed: 12/23/2022] Open
Abstract
Background: The aim of the study was to evaluate the role of different immunohistochemical and radiomics features in patients with small cell lung cancer (SCLC). Methods: Consecutive patients with histologically proven SCLC with limited (n = 47, 48%) or extensive disease (n = 51, 52%) treated with radiotherapy and chemotherapy at our department were included in the analysis. The expression of different immunohistochemical markers from the initial tissue biopsy, such as CD56, CD44, chromogranin A, synaptophysin, TTF-1, GLUT-1, Hif-1 a, PD-1, and PD-L1, and MIB-1/KI-67 as well as LDH und NSE from the initial blood sample were evaluated. H-scores were additionally generated for CD44, Hif-1a, and GLUT-1. A total of 72 computer tomography (CT) radiomics texture features from a homogenous subgroup (n = 31) of patients were correlated with the immunohistochemistry, the survival (OS), and the progression-free survival (PFS). Results: The median OS, calculated from diagnosis, was 21 months for patients with limited disease and 13 months for patients with extensive disease. The expression of synaptophysin correlated with a better OS (HR 0.546 95% CI 0.308–0.966, p = 0.03). The expression of TTF-1 (HR 0.286, 95% CI: 0.117–0.698, p = 0.006) and a lower GLUT-1 H-score (median = 50, HR: 0.511, 95% CI: 0.260–1.003, p = 0.05) correlated with a better PFS. Patients without chromogranin A expression had a higher risk for developing cerebral metastases (p = 0.02) and patients with PD 1 expression were at risk for developing metastases (p = 0.02). Our radiomics analysis did not reveal a single texture feature that correlated highly with OS or PFS. Correlation coefficients ranged between −0.48 and 0.39 for OS and between −0.46 and 0.38 for PFS. Conclusions: The role of synaptophysin should be further evaluated as synaptophysin-negative patients might profit from treatment intensification. We report an, at most, moderate correlation of radiomics features with overall and progression free survival and no correlation with the expression of different immunohistochemical markers.
Collapse
Affiliation(s)
- Eleni Gkika
- Department of Radiation Oncology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Matthias Benndorf
- Department of Radiology Freiburg, University Medical Center Freiburg, Freiburg, Germany
| | - Benedict Oerther
- Department of Radiology Freiburg, University Medical Center Freiburg, Freiburg, Germany
| | - Farid Mohammad
- Department of Radiology Freiburg, University Medical Center Freiburg, Freiburg, Germany
| | - Susanne Beitinger
- Department of Neurology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Sonja Adebahr
- Department of Radiation Oncology, Medical Center, University Hospital Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Montserrat Carles
- Department of Radiation Oncology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Tanja Schimek-Jasch
- Department of Radiation Oncology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Björn C Frye
- Department of Pneumology, Medical Center, University Hospital Freiburg, Freiburg, Germany
| | - Fabian Bamberg
- Department of Radiology Freiburg, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius F Waller
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany
| | - Martin Werner
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Pathology, Faculty of Medicine, Medical Center, Institute of Surgical Pathology, University Hospital Freiburg, Freiburg, Germany
| | - Anca L Grosu
- German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ursula Nestle
- German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Gian Kayser
- Department of Pathology, Faculty of Medicine, Medical Center, Institute of Surgical Pathology, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
28
|
Asadollahi R, Britschgi C, Joset P, Oneda B, Schindler D, Meier UR, Rauch A. Severe reaction to radiotherapy provoked by hypomorphic germline mutations in ATM (ataxia-telangiectasia mutated gene). Mol Genet Genomic Med 2020; 8:e1409. [PMID: 32748564 PMCID: PMC7549565 DOI: 10.1002/mgg3.1409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Background A minority of breast cancer (BC) patients suffer from severe reaction to adjuvant radiotherapy (RT). Although deficient DNA double‐strand break repair is considered the main basis for the reactions, pretreatment identification of high‐risk patients has been challenging. Methods To retrospectively determine the etiology of severe local reaction to RT in a 39‐year‐old woman with BC, we performed next‐generation sequencing followed by further clinical and functional studies. Results We found a −4 intronic variant (c.2251‐4A>G) in trans with a synonymous (c.3576G>A) variant affecting the ATM DNA‐repair gene (NG_009830.1, NM_000051.3) which is linked to autosomal recessive ataxia–telangiectasia (A–T). We verified abnormal transcripts resulting from both variants, next to a minor wild‐type transcript leading to a residual ATM kinase activity and genomic instability. Follow‐up examination of the patient revealed no classic sign of A–T but previously unnoticed head dystonia and mild dysarthria, a family history of BC and late‐onset ataxia segregating with the variants. Additionally, her serum level of alpha‐fetoprotein (AFP) was elevated similar to A–T patients. Conclusion Considering the variable presentations of A–T and devastating impact of severe reactions to RT, we suggest a routine measurement of AFP in RT‐candidate BC patients followed by next‐generation sequencing with special attention to non‐canonical splice site and synonymous variants in ATM.
Collapse
Affiliation(s)
- Reza Asadollahi
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Christian Britschgi
- Department of Medical Oncology and Hematology, University Hospital Zurich, Comprehensive Cancer Center Zurich and University of Zurich, Zurich, Switzerland
| | - Pascal Joset
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Beatrice Oneda
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Detlev Schindler
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Urs R Meier
- Department of Radiation Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland.,Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Kang J, Coates JT, Strawderman RL, Rosenstein BS, Kerns SL. Genomics models in radiotherapy: From mechanistic to machine learning. Med Phys 2020; 47:e203-e217. [PMID: 32418335 PMCID: PMC8725063 DOI: 10.1002/mp.13751] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/28/2019] [Accepted: 07/17/2019] [Indexed: 12/28/2022] Open
Abstract
Machine learning (ML) provides a broad framework for addressing high-dimensional prediction problems in classification and regression. While ML is often applied for imaging problems in medical physics, there are many efforts to apply these principles to biological data toward questions of radiation biology. Here, we provide a review of radiogenomics modeling frameworks and efforts toward genomically guided radiotherapy. We first discuss medical oncology efforts to develop precision biomarkers. We next discuss similar efforts to create clinical assays for normal tissue or tumor radiosensitivity. We then discuss modeling frameworks for radiosensitivity and the evolution of ML to create predictive models for radiogenomics.
Collapse
Affiliation(s)
- John Kang
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - James T. Coates
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Robert L. Strawderman
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642, USA
| | - Barry S. Rosenstein
- Department of Radiation Oncology and the Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah L. Kerns
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
30
|
Wu J, Sheng VS, Zhang J, Li H, Dadakova T, Swisher CL, Cui Z, Zhao P. Multi-Label Active Learning Algorithms for Image Classification: Overview and Future Promise. ACM COMPUTING SURVEYS 2020; 53:28. [PMID: 34421185 PMCID: PMC8376181 DOI: 10.1145/3379504] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 12/01/2019] [Indexed: 05/13/2023]
Abstract
Image classification is a key task in image understanding, and multi-label image classification has become a popular topic in recent years. However, the success of multi-label image classification is closely related to the way of constructing a training set. As active learning aims to construct an effective training set through iteratively selecting the most informative examples to query labels from annotators, it was introduced into multi-label image classification. Accordingly, multi-label active learning is becoming an important research direction. In this work, we first review existing multi-label active learning algorithms for image classification. These algorithms can be categorized into two top groups from two aspects respectively: sampling and annotation. The most important component of multi-label active learning is to design an effective sampling strategy that actively selects the examples with the highest informativeness from an unlabeled data pool, according to various information measures. Thus, different informativeness measures are emphasized in this survey. Furthermore, this work also makes a deep investigation on existing challenging issues and future promises in multi-label active learning with a focus on four core aspects: example dimension, label dimension, annotation, and application extension.
Collapse
Affiliation(s)
- Jian Wu
- Soochow University, China and Human Longevity, Inc., USA
| | | | - Jing Zhang
- Nanjing University of Science and Technology, China
| | - Hua Li
- Washington University in St. Louis, USA
| | | | | | - Zhiming Cui
- Suzhou University of Science and Technology, China
| | | |
Collapse
|
31
|
Samaga D, Hornung R, Braselmann H, Hess J, Zitzelsberger H, Belka C, Boulesteix AL, Unger K. Single-center versus multi-center data sets for molecular prognostic modeling: a simulation study. Radiat Oncol 2020; 15:109. [PMID: 32410693 PMCID: PMC7227093 DOI: 10.1186/s13014-020-01543-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background Prognostic models based on high-dimensional omics data generated from clinical patient samples, such as tumor tissues or biopsies, are increasingly used for prognosis of radio-therapeutic success. The model development process requires two independent discovery and validation data sets. Each of them may contain samples collected in a single center or a collection of samples from multiple centers. Multi-center data tend to be more heterogeneous than single-center data but are less affected by potential site-specific biases. Optimal use of limited data resources for discovery and validation with respect to the expected success of a study requires dispassionate, objective decision-making. In this work, we addressed the impact of the choice of single-center and multi-center data as discovery and validation data sets, and assessed how this impact depends on the three data characteristics signal strength, number of informative features and sample size. Methods We set up a simulation study to quantify the predictive performance of a model trained and validated on different combinations of in silico single-center and multi-center data. The standard bioinformatical analysis workflow of batch correction, feature selection and parameter estimation was emulated. For the determination of model quality, four measures were used: false discovery rate, prediction error, chance of successful validation (significant correlation of predicted and true validation data outcome) and model calibration. Results In agreement with literature about generalizability of signatures, prognostic models fitted to multi-center data consistently outperformed their single-center counterparts when the prediction error was the quality criterion of interest. However, for low signal strengths and small sample sizes, single-center discovery sets showed superior performance with respect to false discovery rate and chance of successful validation. Conclusions With regard to decision making, this simulation study underlines the importance of study aims being defined precisely a priori. Minimization of the prediction error requires multi-center discovery data, whereas single-center data are preferable with respect to false discovery rate and chance of successful validation when the expected signal or sample size is low. In contrast, the choice of validation data solely affects the quality of the estimator of the prediction error, which was more precise on multi-center validation data.
Collapse
Affiliation(s)
- Daniel Samaga
- Helmholtz Zentrum, München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.
| | - Roman Hornung
- Department of Medical Information Processing, Biometry and Epidemiology, University of Munich, Marchioninistr. 15, Munich, 81377, Germany
| | - Herbert Braselmann
- Helmholtz Zentrum, München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany
| | - Julia Hess
- Helmholtz Zentrum, München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum München, Research Center for Environmental Health (GmbH), Munich, Ingolstädter Landstr. 1, Munich, 85764, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, Munich, 81377, Germany
| | - Horst Zitzelsberger
- Helmholtz Zentrum, München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum München, Research Center for Environmental Health (GmbH), Munich, Ingolstädter Landstr. 1, Munich, 85764, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, Munich, 81377, Germany
| | - Claus Belka
- Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum München, Research Center for Environmental Health (GmbH), Munich, Ingolstädter Landstr. 1, Munich, 85764, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, Munich, 81377, Germany
| | - Anne-Laure Boulesteix
- Department of Medical Information Processing, Biometry and Epidemiology, University of Munich, Marchioninistr. 15, Munich, 81377, Germany
| | - Kristian Unger
- Helmholtz Zentrum, München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum München, Research Center for Environmental Health (GmbH), Munich, Ingolstädter Landstr. 1, Munich, 85764, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, Munich, 81377, Germany
| |
Collapse
|
32
|
An FTIR Microspectroscopy Ratiometric Approach for Monitoring X-ray Irradiation Effects on SH-SY5Y Human Neuroblastoma Cells. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10082974] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability of Fourier transform infrared (FTIR) spectroscopy in analyzing cells at a molecular level was exploited for investigating the biochemical changes induced in protein, nucleic acid, lipid, and carbohydrate content of cells after irradiation by graded X-ray doses. Infrared spectra from in vitro SH-SY5Y neuroblastoma cells following exposure to X-rays (0, 2, 4, 6, 8, 10 Gy) were analyzed using a ratiometric approach by evaluating the ratios between the absorbance of significant peaks. The spectroscopic investigation was performed on cells fixed immediately (t0 cells) and 24 h (t24 cells) after irradiation to study both the initial radiation-induced damage and the effect of the ensuing cellular repair processes. The analysis of infrared spectra allowed us to detect changes in proteins, lipids, and nucleic acids attributable to X-ray exposure. The ratiometric analysis was able to quantify changes for the protein, lipid, and DNA components and to suggest the occurrence of apoptosis processes. The ratiometric study of Amide I band indicated also that the secondary structure of proteins was significantly modified. The comparison between the results from t0 and t24 cells indicated the occurrence of cellular recovery processes. The adopted approach can provide a very direct way to monitor changes for specific cellular components and can represent a valuable tool for developing innovative strategies to monitor cancer radiotherapy outcome.
Collapse
|
33
|
Averbeck D, Candéias S, Chandna S, Foray N, Friedl AA, Haghdoost S, Jeggo PA, Lumniczky K, Paris F, Quintens R, Sabatier L. Establishing mechanisms affecting the individual response to ionizing radiation. Int J Radiat Biol 2020; 96:297-323. [PMID: 31852363 DOI: 10.1080/09553002.2019.1704908] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose: Humans are increasingly exposed to ionizing radiation (IR). Both low (<100 mGy) and high doses can cause stochastic effects, including cancer; whereas doses above 100 mGy are needed to promote tissue or cell damage. 10-15% of radiotherapy (RT) patients suffer adverse reactions, described as displaying radiosensitivity (RS). Sensitivity to IR's stochastic effects is termed radiosusceptibility (RSu). To optimize radiation protection we need to understand the range of individual variability and underlying mechanisms. We review the potential mechanisms contributing to RS/RSu focusing on RS following RT, the most tractable RS group.Conclusions: The IR-induced DNA damage response (DDR) has been well characterized. Patients with mutations in the DDR have been identified and display marked RS but they represent only a small percentage of the RT patients with adverse reactions. We review the impacting mechanisms and additional factors influencing RS/RSu. We discuss whether RS/RSu might be genetically determined. As a recommendation, we propose that a prospective study be established to assess RS following RT. The study should detail tumor site and encompass a well-defined grading system. Predictive assays should be independently validated. Detailed analysis of the inflammatory, stress and immune responses, mitochondrial function and life style factors should be included. Existing cohorts should also be optimally exploited.
Collapse
Affiliation(s)
| | - Serge Candéias
- CEA, CNRS, LCMB, University of Grenoble Alpes, Grenoble, France
| | - Sudhir Chandna
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Nicolas Foray
- Inserm UA8 Unit Radiations: Defense, Health and Environment, Lyon, France
| | - Anna A Friedl
- Department of Radiation Oncology, University Hospital, LMU, Munich, Germany
| | - Siamak Haghdoost
- Cimap-Laria, Advanced Resource Center for HADrontherapy in Europe (ARCHADE,), University of Caen Normandy, France.,Centre for Radiation Protection Research, Department of Molecular Bioscience, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Penelope A Jeggo
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Katalin Lumniczky
- Department of Radiation Medicine, Division of Radiobiology and Radiohygiene, National Public Health Center, Budapest, Hungary
| | | | | | | |
Collapse
|
34
|
Blakely EA, Faddegon B, Tinkle C, Bloch C, Dominello M, Griffin RJ, Joiner MC, Burmeister J. Three discipline collaborative radiation therapy (3DCRT) special debate: The United States needs at least one carbon ion facility. J Appl Clin Med Phys 2019; 20:6-13. [PMID: 31573146 PMCID: PMC6839391 DOI: 10.1002/acm2.12727] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 01/07/2023] Open
Affiliation(s)
| | - Bruce Faddegon
- Department of Radiation OncologyUniversity of California – San FranciscoSan FranciscoCAUSA
| | - Christopher Tinkle
- Department of Radiation OncologySt. Jude Children’s Research HospitalMemphisTNUSA
| | - Charles Bloch
- Department of Radiation OncologyUniversity of WashingtonSeattleWAUSA
| | - Michael Dominello
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
| | - Robert J Griffin
- Department of OncologyUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Michael C Joiner
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
| | - Jay Burmeister
- Department of OncologyWayne State University School of MedicineDetroitMIUSA,Gershenson Radiation Oncology CenterBarbara Ann Karmanos Cancer InstituteDetroitMIUSA
| |
Collapse
|
35
|
A new facility for proton radiobiology at the Trento proton therapy centre: Design and implementation. Phys Med 2019; 58:99-106. [DOI: 10.1016/j.ejmp.2019.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 01/26/2023] Open
|