1
|
Zhu Y, Li Z, Wu Z, Zhuo T, Dai L, Liang G, Peng H, Lu H, Wang Y. MIS18A upregulation promotes cell viability, migration and tumor immune evasion in lung adenocarcinoma. Oncol Lett 2024; 28:376. [PMID: 38910901 PMCID: PMC11190817 DOI: 10.3892/ol.2024.14509] [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: 02/03/2024] [Accepted: 05/13/2024] [Indexed: 06/25/2024] Open
Abstract
Lung adenocarcinoma (LUAD) presents a significant global health challenge owing to its poor prognosis and high mortality rates. Despite its involvement in the initiation and progression of a number of cancer types, the understanding of the precise impact of MIS18 kinetochore protein A (MIS18A) on LUAD remains incomplete. In the present study, the role of MIS18A in LUAD was investigated by analyzing the genomic and clinical data from multiple public datasets. The expression of MIS18A was validated using reverse transcription-quantitative polymerase chain reaction, and in vitro experiments involving small interfering RNA-induced downregulation of MIS18A in lung cancer cells were conducted to further explore its impact. These findings revealed that elevated MIS18A expression in LUAD was associated with advanced clinical features and poor prognosis. Functional analysis also revealed the role of MIS18A in regulating the cell cycle and immune-related pathways. Moreover, MIS18A altered the immune microenvironment in LUAD, influencing its response to immunotherapy and drug sensitivity. The results of the in vitro experiments indicated that suppression of MIS18A expression reduced the proliferative and migratory capacities of LUAD cells. In summary, MIS18A possesses potential as a biomarker and may serve as a possible therapeutic target for LUAD, with significant implications for tumor progression by influencing both cell cycle dynamics and immune infiltration.
Collapse
Affiliation(s)
- Yongjie Zhu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zihao Li
- Department of Thoracic Surgery, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi Zhuang Autonomous Region 545026, P.R. China
| | - Zuotao Wu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Ting Zhuo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Lei Dai
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guanbiao Liang
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Huajian Peng
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Honglin Lu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yongyong Wang
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
2
|
Ren F, Fei Q, Qiu K, Zhang Y, Zhang H, Sun L. Liquid biopsy techniques and lung cancer: diagnosis, monitoring and evaluation. J Exp Clin Cancer Res 2024; 43:96. [PMID: 38561776 PMCID: PMC10985944 DOI: 10.1186/s13046-024-03026-7] [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: 01/12/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Lung cancer stands as the most prevalent form of cancer globally, posing a significant threat to human well-being. Due to the lack of effective and accurate early diagnostic methods, many patients are diagnosed with advanced lung cancer. Although surgical resection is still a potential means of eradicating lung cancer, patients with advanced lung cancer usually miss the best chance for surgical treatment, and even after surgical resection patients may still experience tumor recurrence. Additionally, chemotherapy, the mainstay of treatment for patients with advanced lung cancer, has the potential to be chemo-resistant, resulting in poor clinical outcomes. The emergence of liquid biopsies has garnered considerable attention owing to their noninvasive nature and the ability for continuous sampling. Technological advancements have propelled circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), tumor metabolites, tumor-educated platelets (TEPs), and tumor-associated antigens (TAA) to the forefront as key liquid biopsy biomarkers, demonstrating intriguing and encouraging results for early diagnosis and prognostic evaluation of lung cancer. This review provides an overview of molecular biomarkers and assays utilized in liquid biopsies for lung cancer, encompassing CTCs, ctDNA, non-coding RNA (ncRNA), EVs, tumor metabolites, TAAs and TEPs. Furthermore, we expound on the practical applications of liquid biopsies, including early diagnosis, treatment response monitoring, prognostic evaluation, and recurrence monitoring in the context of lung cancer.
Collapse
Affiliation(s)
- Fei Ren
- Department of Geriatrics, The First Hospital of China Medical University, Shen Yang, 110000, China
| | - Qian Fei
- Department of Oncology, Shengjing Hospital of China Medical University, Shen Yang, 110000, China
| | - Kun Qiu
- Thoracic Surgery, The First Hospital of China Medical University, Shen Yang, 110000, China
| | - Yuanjie Zhang
- Thoracic Surgery, The First Hospital of China Medical University, Shen Yang, 110000, China
| | - Heyang Zhang
- Department of Hematology, The First Hospital of China Medical University, Shen Yang, 110000, China.
| | - Lei Sun
- Thoracic Surgery, The First Hospital of China Medical University, Shen Yang, 110000, China.
| |
Collapse
|
3
|
Fiste O, Gkiozos I, Charpidou A, Syrigos NK. Artificial Intelligence-Based Treatment Decisions: A New Era for NSCLC. Cancers (Basel) 2024; 16:831. [PMID: 38398222 PMCID: PMC10887017 DOI: 10.3390/cancers16040831] [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: 01/31/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality among women and men, in developed countries, despite the public health interventions including tobacco-free campaigns, screening and early detection methods, recent therapeutic advances, and ongoing intense research on novel antineoplastic modalities. Targeting oncogenic driver mutations and immune checkpoint inhibition has indeed revolutionized NSCLC treatment, yet there still remains the unmet need for robust and standardized predictive biomarkers to accurately inform clinical decisions. Artificial intelligence (AI) represents the computer-based science concerned with large datasets for complex problem-solving. Its concept has brought a paradigm shift in oncology considering its immense potential for improved diagnosis, treatment guidance, and prognosis. In this review, we present the current state of AI-driven applications on NSCLC management, with a particular focus on radiomics and pathomics, and critically discuss both the existing limitations and future directions in this field. The thoracic oncology community should not be discouraged by the likely long road of AI implementation into daily clinical practice, as its transformative impact on personalized treatment approaches is undeniable.
Collapse
Affiliation(s)
- Oraianthi Fiste
- Oncology Unit, Third Department of Internal Medicine and Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (A.C.); (N.K.S.)
| | | | | | | |
Collapse
|
4
|
Wang J, Wang J, Huang X, Zhou Y, Qi J, Sun X, Nie J, Hu Z, Wang S, Hong B, Wang H. CT radiomics-based model for predicting TMB and immunotherapy response in non-small cell lung cancer. BMC Med Imaging 2024; 24:45. [PMID: 38360550 PMCID: PMC10870537 DOI: 10.1186/s12880-024-01221-8] [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: 10/11/2023] [Accepted: 02/03/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Tumor mutational burden (TMB) is one of the most significant predictive biomarkers of immunotherapy efficacy in non-small cell lung cancer (NSCLC). Radiomics allows high-throughput extraction and analysis of advanced and quantitative medical imaging features. This study develops and validates a radiomic model for predicting TMB level and the response to immunotherapy based on CT features in NSCLC. METHOD Pre-operative chest CT images of 127 patients with NSCLC were retrospectively studied. The 3D-Slicer software was used to outline the region of interest and extract features from the CT images. Radiomics prediction model was constructed by LASSO and multiple logistic regression in a training dataset. The model was validated by receiver operating characteristic (ROC) curves and calibration curves using external datasets. Decision curve analysis was used to assess the value of the model for clinical application. RESULTS A total of 1037 radiomic features were extracted from the CT images of NSCLC patients from TCGA. LASSO regression selected three radiomics features (Flatness, Autocorrelation and Minimum), which were associated with TMB level in NSCLC. A TMB prediction model consisting of 3 radiomic features was constructed by multiple logistic regression. The area under the curve (AUC) value in the TCGA training dataset was 0.816 (95% CI: 0.7109-0.9203) for predicting TMB level in NSCLC. The AUC value in external validation dataset I was 0.775 (95% CI: 0.5528-0.9972) for predicting TMB level in NSCLC, and the AUC value in external validation dataset II was 0.762 (95% CI: 0.5669-0.9569) for predicting the efficacy of immunotherapy in NSCLC. CONCLUSION The model based on CT radiomic features helps to achieve cost effective improvement in TMB classification and precise immunotherapy treatment of NSCLC patients.
Collapse
Affiliation(s)
- Jiexiao Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jialiang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Xiang Huang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Yanfei Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jian Qi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaojun Sun
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Jinfu Nie
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Zongtao Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Shujie Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.
| | - Bo Hong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China.
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China.
| |
Collapse
|
5
|
Boopathy LK, Roy A, Gopal T, Kandy RRK, Arumugam MK. Potential molecular mechanisms of myrtenal against colon cancer: A systematic review. J Biochem Mol Toxicol 2024; 38:e23525. [PMID: 37665681 DOI: 10.1002/jbt.23525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023]
Abstract
Colon cancer is a serious health problem across the globe with various dietary lifestyle modifications. It arises as an inflammation mediated crypts in the colon epithelial cells and undergoes uncontrolled cell division and proliferation. Bacterial enzymes contribute to a major outbreak in colon cancer development upon the release of toxic metabolites from the gut microflora. Pathogen associated molecular patterns and damage associated molecular patterns triggers the NLPR3 inflammasome pathways that releases pro-inflammatory cytokines to induce cancer of the colon. Contributing to this, specific chemokines and receptor complexes attribute to cellular proliferation and metastasis. Bacterial enzymes synergistically attack the colon mucosa and degenerate the cellular integrity causing lysosomal discharge. These factors further instigate the Tol like receptors (TLRs) and Nod like receptors (NLRs) to promote angiogenesis and supply nutrients for the cancer cells. Myrtenal, a monoterpene, is gaining more importance in recent times and it is being widely utilized against many diseases such as cancers, neurodegenerative diseases and diabetes. Based on the research data's, the reviews focus on the anticancer property of myrtenal by emphasizing its therapeutic properties which downregulate the inflammasome pathways and other signalling pathways. Combination therapy is gaining more importance as they can target every variant in the cellular stress condition. Clinical studies with compounds like myrtenal of the monoterpenes family is provided with positive results which might open an effective anticancer drug therapy. This review highlights myrtenal and its biological potency as a cost effective drug for prevention and treatment of colon cancer.
Collapse
Affiliation(s)
- Lokesh Kumar Boopathy
- Centre for Laboratory Animal Technology and Research, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Anitha Roy
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Thiyagarajan Gopal
- Centre for Laboratory Animal Technology and Research, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rakhee Rathnam Kalari Kandy
- Department of Biochemistry and Molecular Biology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Madan Kumar Arumugam
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
6
|
Derbal Y. Adaptive Control of Tumor Growth. Cancer Control 2024; 31:10732748241230869. [PMID: 38294947 PMCID: PMC10832444 DOI: 10.1177/10732748241230869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/04/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
Cancer treatment optimizations select the most optimum combinations of drugs, sequencing schedules, and appropriate doses that would limit toxicity and yield an improved patient quality of life. However, these optimizations often lack an adequate consideration of cancer's near-infinite potential for evolutionary adaptation to therapeutic interventions. Adapting cancer therapy based on monitored tumor burden and clonal composition is an intuitively sound approach to the treatment of cancer as an inherently complex and adaptive system. The adaptation would be driven by clinical outcome setpoints embodying the aims to thwart therapeutic resistance and maintain a long-term management of the disease or even a cure. However, given the nonlinear, stochastic dynamics of tumor response to therapeutic interventions, adaptive therapeutic strategies may at least need a one-step-ahead prediction of tumor burden to maintain their control over tumor growth dynamics. The article explores the feasibility of adaptive cancer treatment driven by tumor state feedback assuming cell adaptive fitness to be the underlying source of phenotypic plasticity and pathway entropy as a biomarker of tumor growth trajectory. The exploration is undertaken using deterministic and stochastic models of tumor growth dynamics.
Collapse
Affiliation(s)
- Youcef Derbal
- Ted Rogers School of Information Technology Management, Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
7
|
Derbal Y. Adaptive Cancer Therapy in the Age of Generative Artificial Intelligence. Cancer Control 2024; 31:10732748241264704. [PMID: 38897721 PMCID: PMC11189021 DOI: 10.1177/10732748241264704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
Therapeutic resistance is a major challenge facing the design of effective cancer treatments. Adaptive cancer therapy is in principle the most viable approach to manage cancer's adaptive dynamics through drug combinations with dose timing and modulation. However, there are numerous open issues facing the clinical success of adaptive therapy. Chief among these issues is the feasibility of real-time predictions of treatment response which represent a bedrock requirement of adaptive therapy. Generative artificial intelligence has the potential to learn prediction models of treatment response from clinical, molecular, and radiomics data about patients and their treatments. The article explores this potential through a proposed integration model of Generative Pre-Trained Transformers (GPTs) in a closed loop with adaptive treatments to predict the trajectories of disease progression. The conceptual model and the challenges facing its realization are discussed in the broader context of artificial intelligence integration in oncology.
Collapse
Affiliation(s)
- Youcef Derbal
- Ted Rogers School of Information Technology Management, Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
8
|
Rinaldi L, Guerini Rocco E, Spitaleri G, Raimondi S, Attili I, Ranghiero A, Cammarata G, Minotti M, Lo Presti G, De Piano F, Bellerba F, Funicelli G, Volpe S, Mora S, Fodor C, Rampinelli C, Barberis M, De Marinis F, Jereczek-Fossa BA, Orecchia R, Rizzo S, Botta F. Association between Contrast-Enhanced Computed Tomography Radiomic Features, Genomic Alterations and Prognosis in Advanced Lung Adenocarcinoma Patients. Cancers (Basel) 2023; 15:4553. [PMID: 37760521 PMCID: PMC10527057 DOI: 10.3390/cancers15184553] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Non-invasive methods to assess mutational status, as well as novel prognostic biomarkers, are warranted to foster therapy personalization of patients with advanced non-small cell lung cancer (NSCLC). This study investigated the association of contrast-enhanced Computed Tomography (CT) radiomic features of lung adenocarcinoma lesions, alone or integrated with clinical parameters, with tumor mutational status (EGFR, KRAS, ALK alterations) and Overall Survival (OS). In total, 261 retrospective and 48 prospective patients were enrolled. A Radiomic Score (RS) was created with LASSO-Logistic regression models to predict mutational status. Radiomic, clinical and clinical-radiomic models were trained on retrospective data and tested (Area Under the Curve, AUC) on prospective data. OS prediction models were trained and tested on retrospective data with internal cross-validation (C-index). RS significantly predicted each alteration at training (radiomic and clinical-radiomic AUC 0.95-0.98); validation performance was good for EGFR (AUC 0.86), moderate for KRAS and ALK (AUC 0.61-0.65). RS was also associated with OS at univariate and multivariable analysis, in the latter with stage and type of treatment. The validation C-index was 0.63, 0.79, and 0.80 for clinical, radiomic, and clinical-radiomic models. The study supports the potential role of CT radiomics for non-invasive identification of gene alterations and prognosis prediction in patients with advanced lung adenocarcinoma, to be confirmed with independent studies.
Collapse
Affiliation(s)
- Lisa Rinaldi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| | - Elena Guerini Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Alberto Ranghiero
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Giulio Cammarata
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Marta Minotti
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Giuliana Lo Presti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Francesca De Piano
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Federica Bellerba
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Gianluigi Funicelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Stefania Volpe
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Serena Mora
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiana Fodor
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiano Rampinelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Massimo Barberis
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Roberto Orecchia
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
- Scientific Direction, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), Via Tesserete 46, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Via G. Buffi 13, 6900 Lugano, Switzerland
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| |
Collapse
|
9
|
Ferrante M, Rinaldi L, Botta F, Hu X, Dolp A, Minotti M, De Piano F, Funicelli G, Volpe S, Bellerba F, De Marco P, Raimondi S, Rizzo S, Shi K, Cremonesi M, Jereczek-Fossa BA, Spaggiari L, De Marinis F, Orecchia R, Origgi D. Application of nnU-Net for Automatic Segmentation of Lung Lesions on CT Images and Its Implication for Radiomic Models. J Clin Med 2022; 11:7334. [PMID: 36555950 PMCID: PMC9784875 DOI: 10.3390/jcm11247334] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Radiomics investigates the predictive role of quantitative parameters calculated from radiological images. In oncology, tumour segmentation constitutes a crucial step of the radiomic workflow. Manual segmentation is time-consuming and prone to inter-observer variability. In this study, a state-of-the-art deep-learning network for automatic segmentation (nnU-Net) was applied to computed tomography images of lung tumour patients, and its impact on the performance of survival radiomic models was assessed. In total, 899 patients were included, from two proprietary and one public datasets. Different network architectures (2D, 3D) were trained and tested on different combinations of the datasets. Automatic segmentations were compared to reference manual segmentations performed by physicians using the DICE similarity coefficient. Subsequently, the accuracy of radiomic models for survival classification based on either manual or automatic segmentations were compared, considering both hand-crafted and deep-learning features. The best agreement between automatic and manual contours (DICE = 0.78 ± 0.12) was achieved averaging 2D and 3D predictions and applying customised post-processing. The accuracy of the survival classifier (ranging between 0.65 and 0.78) was not statistically different when using manual versus automatic contours, both with hand-crafted and deep features. These results support the promising role nnU-Net can play in automatic segmentation, accelerating the radiomic workflow without impairing the models' accuracy. Further investigations on different clinical endpoints and populations are encouraged to confirm and generalise these findings.
Collapse
Affiliation(s)
- Matteo Ferrante
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Lisa Rinaldi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Xiaobin Hu
- Department of Informatics, Technical University of Munich, Arcisstraße 21, 80333 Munich, Germany
| | - Andreas Dolp
- Department of Informatics, Technical University of Munich, Arcisstraße 21, 80333 Munich, Germany
| | - Marta Minotti
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Francesca De Piano
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Gianluigi Funicelli
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Volpe
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Federica Bellerba
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), via Tesserete 46, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), via G. Buffi 13, 6900 Lugano, Switzerland
| | - Kuangyu Shi
- Chair for Computer-Aided Medical Procedures, Department of Informatics, Technical University of Munich, Arcisstraße 21, 80333 Munich, Germany
- Department of Nuclear Medicine, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Marta Cremonesi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Barbara A. Jereczek-Fossa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Lorenzo Spaggiari
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
- Division of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Roberto Orecchia
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
- Scientific Direction, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
10
|
Han X, Tang X, Zhu H, Zhu D, Zhang X, Meng X, Hua Y, Wang Z, Zhang Y, Huang W, Wang L, Yuan S, Zhang P, Gong H, Sun Y, Zhang Y, Liu Z, Dong X, Gai F, Huang Z, Zhu C, Guo J, Wang Z. Short-term dynamics of circulating tumor DNA predicting efficacy of sintilimab plus docetaxel in second-line treatment of advanced NSCLC: biomarker analysis from a single-arm, phase 2 trial. J Immunother Cancer 2022; 10:jitc-2022-004952. [PMID: 36600554 PMCID: PMC9730395 DOI: 10.1136/jitc-2022-004952] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Robust biomarker predicting efficacy of immunotherapy is limited. Circulating tumor DNA (ctDNA) sought to effectively monitor therapeutic response as well as disease progression. This study aims to investigate predictive role of ctDNA short-term dynamic change (6 weeks postimmunotherapy) in a single-arm, phase 2 trial of sintilimab plus docetaxel for previously treated advanced non-small cell lung cancer (NSCLC) patients. METHODS A total of 33 patients with advanced NSCLC with disease progression during or after any first-line treatment were prospectively enrolled between 2019 and 2020. Patients received sintilimab (200 mg, day 1, every 3 weeks) plus docetaxel (75 mg/m2, day 3, every 3 weeks) for 4-6 cycles, followed by maintenance therapy with sintilimab (200 mg, day 1, every 3 weeks) until disease progression or unacceptable toxic effects. Blood samples were prospectively collected at baseline, and after 2 cycles of treatment (6 weeks post-treatment). All samples were subjected to targeted next-generation sequencing with a panel of 448 cancer-related genes. The landscape of high-frequency genomic profile of baseline and 6th week was described. Major molecular characteristics in preselected genes of interest associated with response to second-line chemoimmunotherapy were analyzed. The curative effects and prognosis of patients were evaluated. RESULTS Patients with ctDNA clearance at 6th week had decreased tumor volume, while most patients with positive ctDNA at 6th-week experienced an increase in tumor volume. Positive 6th-week ctDNA was associated with significantly shorter progression-free survival (PFS) (91 vs NR days; p<0.0001) and overall survival (47 vs 467 days; p =0.0039). Clearance of clonal mutations and none new clonal formation at 6th week were associated with longer PFS (mPFS 89 vs 266 days, p =0.003). ctDNA clearance at 6th week was an independent risk factor for progression or death (HR=100 (95% CI 4.10 to 2503.00), p=0.005). CONCLUSION ctDNA status and ctDNA mutation clearance putatively serve as predictive biomarkers for sintilimab combined with docetaxel chemotherapy in pretreated advanced NSCLC patients.
Collapse
Affiliation(s)
- Xiao Han
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Xiaoyong Tang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Hui Zhu
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Dongyuan Zhu
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Xiqin Zhang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ying Hua
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Zhongtang Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Yan Zhang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Wei Huang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Linlin Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Shuanghu Yuan
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Pinliang Zhang
- Internal Medicine Department, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Heyi Gong
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Yulan Sun
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Yingjie Zhang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Zengjun Liu
- Internal Medicine Department, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Xiaomeng Dong
- Medical Department, Amoy Diagnostics Co Ltd, Xiamen, Fujian, China
| | - Fei Gai
- Medical Department, Amoy Diagnostics Co Ltd, Xiamen, Fujian, China
| | - Zhan Huang
- Medical Department, Amoy Diagnostics Co Ltd, Xiamen, Fujian, China
| | - Changbin Zhu
- Medical Department, Amoy Diagnostics Co Ltd, Xiamen, Fujian, China
| | - Jun Guo
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Zhehai Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| |
Collapse
|
11
|
Blee JA, Liu X, Harland AJ, Fatania K, Currie S, Kurian KM, Hauert S. Liquid biopsies for early diagnosis of brain tumours: in silico mathematical biomarker modelling. J R Soc Interface 2022; 19:20220180. [PMID: 35919979 PMCID: PMC9346349 DOI: 10.1098/rsif.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/07/2022] [Indexed: 11/12/2022] Open
Abstract
Brain tumours are the biggest cancer killer in those under 40 and reduce life expectancy more than any other cancer. Blood-based liquid biopsies may aid early diagnosis, prediction and prognosis for brain tumours. It remains unclear whether known blood-based biomarkers, such as glial fibrillary acidic protein (GFAP), have the required sensitivity and selectivity. We have developed a novel in silico model which can be used to assess and compare blood-based liquid biopsies. We focused on GFAP, a putative biomarker for astrocytic tumours and glioblastoma multi-formes (GBMs). In silico modelling was paired with experimental measurement of cell GFAP concentrations and used to predict the tumour volumes and identify key parameters which limit detection. The average GBM volumes of 449 patients at Leeds Teaching Hospitals NHS Trust were also measured and used as a benchmark. Our model predicts that the currently proposed GFAP threshold of 0.12 ng ml-1 may not be suitable for early detection of GBMs, but that lower thresholds may be used. We found that the levels of GFAP in the blood are related to tumour characteristics, such as vasculature damage and rate of necrosis, which are biological markers of tumour aggressiveness. We also demonstrate how these models could be used to provide clinical insight.
Collapse
Affiliation(s)
- Johanna A. Blee
- Department of Engineering Mathematics, University of Bristol, Ada Lovelace Building, Bristol BS8 1TW, UK
| | - Xia Liu
- Brain Tumour Research Centre, Bristol Medical School, Bristol BS2 8DZ, UK
| | - Abigail J. Harland
- Brain Tumour Research Centre, Bristol Medical School, Bristol BS2 8DZ, UK
| | - Kavi Fatania
- Department of Radiology, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - Stuart Currie
- Department of Radiology, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | | | - Sabine Hauert
- Department of Engineering Mathematics, University of Bristol, Ada Lovelace Building, Bristol BS8 1TW, UK
| |
Collapse
|
12
|
EGFR signaling pathway as therapeutic target in human cancers. Semin Cancer Biol 2022; 85:253-275. [PMID: 35427766 DOI: 10.1016/j.semcancer.2022.04.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/12/2022] [Accepted: 04/04/2022] [Indexed: 02/08/2023]
Abstract
Epidermal Growth Factor Receptor (EGFR) enacts major roles in the maintenance of epithelial tissues. However, when EGFR signaling is altered, it becomes the grand orchestrator of epithelial transformation, and hence one of the most world-wide studied tyrosine kinase receptors involved in neoplasia, in several tissues. In the last decades, EGFR-targeted therapies shaped the new era of precision-oncology. Despite major advances, the dream of converting solid tumors into a chronic disease is still unfulfilled, and long-term remission eludes us. Studies investigating the function of this protein in solid malignancies have revealed numerous ways how tumor cells dysregulate EGFR function. Starting from preclinical models (cell lines, organoids, murine models) and validating in clinical specimens, EGFR-related oncogenic pathways, mechanisms of resistance, and novel avenues to inhibit tumor growth and metastatic spread enriching the therapeutic portfolios, were identified. Focusing on non-small cell lung cancer (NSCLC), where EGFR mutations are major players in the adenocarcinoma subtype, we will go over the most relevant discoveries that led us to understand EGFR and beyond, and highlight how they revolutionized cancer treatment by expanding the therapeutic arsenal at our disposal.
Collapse
|
13
|
Treatment-driven tumour heterogeneity and drug resistance: lessons from solid tumours. Cancer Treat Rev 2022; 104:102340. [DOI: 10.1016/j.ctrv.2022.102340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
14
|
Yi Z, Long L, Zeng Y, Liu Z. Current Advances and Challenges in Radiomics of Brain Tumors. Front Oncol 2021; 11:732196. [PMID: 34722274 PMCID: PMC8551958 DOI: 10.3389/fonc.2021.732196] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Imaging diagnosis is crucial for early detection and monitoring of brain tumors. Radiomics enable the extraction of a large mass of quantitative features from complex clinical imaging arrays, and then transform them into high-dimensional data which can subsequently be mined to find their relevance with the tumor's histological features, which reflect underlying genetic mutations and malignancy, along with grade, progression, therapeutic effect, or even overall survival (OS). Compared to traditional brain imaging, radiomics provides quantitative information linked to meaningful biologic characteristics and application of deep learning which sheds light on the full automation of imaging diagnosis. Recent studies have shown that radiomics' application is broad in identifying primary tumor, differential diagnosis, grading, evaluation of mutation status and aggression, prediction of treatment response and recurrence in pituitary tumors, gliomas, and brain metastases. In this descriptive review, besides establishing a general understanding among protocols, results, and clinical significance of these studies, we further discuss the current limitations along with future development of radiomics.
Collapse
Affiliation(s)
- Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lifu Long
- XiangYa School of Medicine, Central South University, Changsha, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Impact of Lesion Delineation and Intensity Quantisation on the Stability of Texture Features from Lung Nodules on CT: A Reproducible Study. Diagnostics (Basel) 2021; 11:diagnostics11071224. [PMID: 34359305 PMCID: PMC8304812 DOI: 10.3390/diagnostics11071224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Computer-assisted analysis of three-dimensional imaging data (radiomics) has received a lot of research attention as a possible means to improve the management of patients with lung cancer. Building robust predictive models for clinical decision making requires the imaging features to be stable enough to changes in the acquisition and extraction settings. Experimenting on 517 lung lesions from a cohort of 207 patients, we assessed the stability of 88 texture features from the following classes: first-order (13 features), Grey-level Co-Occurrence Matrix (24), Grey-level Difference Matrix (14), Grey-level Run-length Matrix (16), Grey-level Size Zone Matrix (16) and Neighbouring Grey-tone Difference Matrix (five). The analysis was based on a public dataset of lung nodules and open-access routines for feature extraction, which makes the study fully reproducible. Our results identified 30 features that had good or excellent stability relative to lesion delineation, 28 to intensity quantisation and 18 to both. We conclude that selecting the right set of imaging features is critical for building clinical predictive models, particularly when changes in lesion delineation and/or intensity quantisation are involved.
Collapse
|