1
|
Rietjens RG, Wang G, van den Berg BM, Rabelink TJ. Spatial metabolomics in tissue injury and regeneration. Curr Opin Genet Dev 2024; 87:102223. [PMID: 38901101 DOI: 10.1016/j.gde.2024.102223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024]
Abstract
Tissue homeostasis is intricately linked to cellular metabolism and metabolite exchange within the tissue microenvironment. The orchestration of adaptive cellular responses during injury and repair depends critically upon metabolic adaptation. This adaptation, in turn, shapes cell fate decisions required for the restoration of tissue homeostasis. Understanding the nuances of metabolic processes within the tissue context and comprehending the intricate communication between cells is therefore imperative for unraveling the complexity of tissue homeostasis and the processes of injury and repair. In this review, we focus on mass spectrometry imaging as an advanced platform with the potential to provide such comprehensive insights into the metabolic instruction governing tissue function. Recent advances in this technology allow to decipher the intricate metabolic networks that determine cellular behavior in the context of tissue resilience, injury, and repair. These insights not only advance our fundamental understanding of tissue biology but also hold implications for therapeutic interventions by targeting metabolic pathways critical for maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Rosalie Gj Rietjens
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine & The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands. https://twitter.com/@RietjensRosalie
| | - Gangqi Wang
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine & The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands. https://twitter.com/@GangqiW
| | - Bernard M van den Berg
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine & The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine & The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
2
|
de Souza N, Zhao S, Bodenmiller B. Multiplex protein imaging in tumour biology. Nat Rev Cancer 2024; 24:171-191. [PMID: 38316945 DOI: 10.1038/s41568-023-00657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
Tissue imaging has become much more colourful in the past decade. Advances in both experimental and analytical methods now make it possible to image protein markers in tissue samples in high multiplex. The ability to routinely image 40-50 markers simultaneously, at single-cell or subcellular resolution, has opened up new vistas in the study of tumour biology. Cellular phenotypes, interaction, communication and spatial organization have become amenable to molecular-level analysis, and application to patient cohorts has identified clinically relevant cellular and tissue features in several cancer types. Here, we review the use of multiplex protein imaging methods to study tumour biology, discuss ongoing attempts to combine these approaches with other forms of spatial omics, and highlight challenges in the field.
Collapse
Affiliation(s)
- Natalie de Souza
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland
- ETH Zurich, Institute of Molecular Systems Biology, Zurich, Switzerland
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Shan Zhao
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Bernd Bodenmiller
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland.
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland.
| |
Collapse
|
3
|
Janßen C, Boskamp T, Le’Clerc Arrastia J, Otero Baguer D, Hauberg-Lotte L, Kriegsmann M, Kriegsmann K, Steinbuß G, Casadonte R, Kriegsmann J, Maaß P. Multimodal Lung Cancer Subtyping Using Deep Learning Neural Networks on Whole Slide Tissue Images and MALDI MSI. Cancers (Basel) 2022; 14:cancers14246181. [PMID: 36551667 PMCID: PMC9776684 DOI: 10.3390/cancers14246181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Artificial intelligence (AI) has shown potential for facilitating the detection and classification of tumors. In patients with non-small cell lung cancer, distinguishing between the most common subtypes, adenocarcinoma (ADC) and squamous cell carcinoma (SqCC), is crucial for the development of an effective treatment plan. This task, however, may still present challenges in clinical routine. We propose a two-modality, AI-based classification algorithm to detect and subtype tumor areas, which combines information from matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) data and digital microscopy whole slide images (WSIs) of lung tissue sections. The method consists of first detecting areas with high tumor cell content by performing a segmentation of the hematoxylin and eosin-stained (H&E-stained) WSIs, and subsequently classifying the tumor areas based on the corresponding MALDI MSI data. We trained the algorithm on six tissue microarrays (TMAs) with tumor samples from N = 232 patients and used 14 additional whole sections for validation and model selection. Classification accuracy was evaluated on a test dataset with another 16 whole sections. The algorithm accurately detected and classified tumor areas, yielding a test accuracy of 94.7% on spectrum level, and correctly classified 15 of 16 test sections. When an additional quality control criterion was introduced, a 100% test accuracy was achieved on sections that passed the quality control (14 of 16). The presented method provides a step further towards the inclusion of AI and MALDI MSI data into clinical routine and has the potential to reduce the pathologist's work load. A careful analysis of the results revealed specific challenges to be considered when training neural networks on data from lung cancer tissue.
Collapse
Affiliation(s)
- Charlotte Janßen
- Center for Industrial Mathematics (ZeTeM), University of Bremen, 28359 Bremen, Germany
- Correspondence:
| | - Tobias Boskamp
- Center for Industrial Mathematics (ZeTeM), University of Bremen, 28359 Bremen, Germany
- Bruker Daltonics, 28359 Bremen, Germany
| | | | - Daniel Otero Baguer
- Center for Industrial Mathematics (ZeTeM), University of Bremen, 28359 Bremen, Germany
| | - Lena Hauberg-Lotte
- Center for Industrial Mathematics (ZeTeM), University of Bremen, 28359 Bremen, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Institute of Pathology Wiesbaden, 65199 Wiesbaden, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Georg Steinbuß
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | - Jörg Kriegsmann
- Proteopath, 54926 Trier, Germany
- Center for Histology, Cytology and Molecular Diagnostic, 54296 Trier, Germany
- Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
| | - Peter Maaß
- Center for Industrial Mathematics (ZeTeM), University of Bremen, 28359 Bremen, Germany
| |
Collapse
|
4
|
Rijcken CJF, De Lorenzi F, Biancacci I, Hanssen RGJM, Thewissen M, Hu Q, Atrafi F, Liskamp RMJ, Mathijssen RHJ, Miedema IHC, Menke-van der Houven van Oordt CW, van Dongen GAMS, Vugts DJ, Timmers M, Hennink WE, Lammers T. Design, development and clinical translation of CriPec®-based core-crosslinked polymeric micelles. Adv Drug Deliv Rev 2022; 191:114613. [PMID: 36343757 DOI: 10.1016/j.addr.2022.114613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Nanomedicines are used to improve the efficacy and safety of pharmacotherapeutic interventions. Unraveling the biological behavior of nanomedicines, including their biodistribution and target site accumulation, is essential to establish design criteria that contribute to superior performance. CriPec® technology is based on amphiphilic methoxy-poly(ethylene glycol)-b-poly[N-(2-hydroxypropyl) methacrylamide lactate] (mPEG-b-pHPMAmLacn) block copolymers, which are designed to upon self-assembly covalently entrap active pharmaceutical ingredients (API) in core-crosslinked polymeric micelles (CCPM). Key features of CCPM are a prolonged circulation time, high concentrations at pathological sites, and low levels of accumulation in the majority of healthy tissues. Proprietary hydrolysable linkers allow for tunable and sustained release of entrapped API, including hydrophobic and hydrophilic small molecules, as well as peptides and oligonucleotides. Preclinical imaging experiments provided valuable information on their tumor and tissue accumulation and distribution, as well as on uptake by cancer, healthy and immune cells. The frontrunner formulation CPC634, which refers to 65 nm-sized CCPM entrapping the chemotherapeutic drug docetaxel, showed excellent pharmacokinetic properties, safety, tumor accumulation and antitumor efficacy in multiple animal models. In the clinic, CPC634 also demonstrated favorable pharmacokinetics, good tolerability, signs of efficacy, and enhanced localization in tumor tissue as compared to conventional docetaxel. PET imaging of radiolabeled CPC634 showed quantifiable accumulation in ∼50 % of tumors and metastases in advanced-stage cancer patients, and demonstrated potential for use in a theranostic setting even when applied at a companion diagnostic dose. Altogether, the preclinical and clinical results obtained to date demonstrate that mPEG-b-pHPMAmLacn CCPM based on CriPec® technology are a potent, tunable, broadly applicable and well-tolerable platform for targeted drug delivery and improved anticancer therapy.
Collapse
Affiliation(s)
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Ilaria Biancacci
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | | | | | - Qizhi Hu
- Cristal Therapeutics, Maastricht, the Netherlands
| | - Florence Atrafi
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Iris H C Miedema
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Matt Timmers
- Cristal Therapeutics, Maastricht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht University, Utrecht, the Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
5
|
Parastar H, Tauler R. Big (Bio)Chemical Data Mining Using Chemometric Methods: A Need for Chemists. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.201801134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hadi Parastar
- Department of Chemistry Sharif University of Technology Tehran Iran
| | - Roma Tauler
- Department of Environmental Chemistry IDAEA-CSIC 08034 Barcelona Spain
| |
Collapse
|
6
|
Wang Z, Zhang Y, Tian R, Luo Z, Zhang R, Li X, Abliz Z. Data-Driven Deciphering of Latent Lesions in Heterogeneous Tissue Using Function-Directed t-SNE of Mass Spectrometry Imaging Data. Anal Chem 2022; 94:13927-13935. [PMID: 36173386 DOI: 10.1021/acs.analchem.2c02990] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mass spectrometry imaging (MSI), which quantifies the underlying chemistry with molecular spatial information in tissue, represents an emerging tool for the functional exploration of pathological progression. Unsupervised machine learning of MSI datasets usually gives an overall interpretation of the metabolic features derived from the abundant ions. However, the features related to the latent lesions are always concealed by the abundant ion features, which hinders precise delineation of the lesions. Herein, we report a data-driven MSI data segmentation approach for recognizing the hidden lesions in the heterogeneous tissue without prior knowledge, which utilizes one-step prediction for feature selection to generate function-specific segmentation maps of the tissue. The performance and robustness of this approach are demonstrated on the MSI datasets of the ischemic rat brain tissues and the human glioma tissue, both possessing different structural complexity and metabolic heterogeneity. Application of the approach to the MSI datasets of the ischemic rat brain tissues reveals the location of the ischemic penumbra, a hidden zone between the ischemic core and the healthy tissue, and instantly discovers the metabolic signatures related to the penumbra. In view of the precise demarcation of latent lesions and the screening of lesion-specific metabolic signatures in tissues, this approach has great potential for in-depth exploration of the metabolic organization of complex tissue.
Collapse
Affiliation(s)
- Zixuan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Yaxin Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Runtao Tian
- Chemmind Technologies Co., Ltd., Beijing 100085, P. R. China
| | - Zhigang Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Ruiping Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Xin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China.,Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, P. R. China
| | - Zeper Abliz
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China.,Center for Imaging and Systems Biology, Minzu University of China, Beijing 100081, P. R. China.,Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, P. R. China
| |
Collapse
|
7
|
Li J, Chen Q, Guo L, Li J, Jin B, Wu X, Shi Y, Xu H, Zheng Y, Wang Y, Du S, Li Z, Lu X, Sang X, Mao Y. In situ Detecting Lipids as Potential Biomarkers for the Diagnosis and Prognosis of Intrahepatic Cholangiocarcinoma. Cancer Manag Res 2022; 14:2903-2912. [PMID: 36187448 PMCID: PMC9524278 DOI: 10.2147/cmar.s357000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/20/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To quantitatively analyze lipid molecules in tumors and adjacent tissues of intrahepatic cholangiocarcinoma (ICC), to establish diagnostic model and to examine lipid changes with clinical classification. Patients and Methods We measured the quantity of 202 lipid molecules in 100 tumor observation points and 100 adjacent observation points of patients who were diagnosed with ICC. Principal component analysis (PCA) and orthogonal partial least squares-discriminant analysis (OPLS-DA) were handles, along with Student’s t-test to identify specific metabolites. Prediction accuracy was validated in the validation set. Another logistic regression model was also established on the training set and validated on the validation set. Results Distinct separation was obtained from PCA and OPLS-DA model. Ten differentiating metabolites were identified using PCA, OPLA-DA and Lasso regression: [m/z 722.5130], [m/z 863.5655], [m/z 436.2834], [m/z 474.2626], [m/z 661.4813], [m/z 750.5443], [m/z 571.2889], [m/z 836.5420], [m/z 772.5862] and [m/z 478.2939]. Using logical regression, a diagnostic equation: y = 3.4*[m/z 436.2834] - 3.773*[m/z 474.2626] + 3.82*[m/z 661.4813] - 4.394*[m/z 863.5655] + 10.165 based on four metabolites successfully differentiated cancerous areas from adjacent normal areas. The AUROC of the model reached 0.993 (95% CI: 0.985–0.999) in the validation group. Compared with the adjacent non-tumor area, three characteristic metabolites FA (22:4), PA (P-18:0/0:0) and Glucosylceramide (d18:1/12:0) showed an increasing trend from stage I to stage II, while seven other metabolites LPA(16:0), PE(34:2), PE(36:4), PE(38:3), PE(40:6), PE(40:5) and LPE(16:0) showed a decreasing trend from stage I to stage II. Conclusion We successfully identified lipid molecules in differentiating tumor tissue and adjacent tissue of ICC, established a discrimination logistic model which could be used as a classifier to classify tumor and non-tumor regions based on analysis in tumor margins and provided information for biomarker changes in ICC, and proposed to related lipid changes with clinical classification.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Qiao Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Lei Guo
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Ji Li
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Xiangan Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Yue Shi
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Yongchang Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Yingyi Wang
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
- Correspondence: Shunda Du; Zhili Li, Email ;
| | - Zhili Li
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| |
Collapse
|
8
|
Prognostic Value of Molecular Intratumor Heterogeneity in Primary Oral Cancer and Its Lymph Node Metastases Assessed by Mass Spectrometry Imaging. Molecules 2022; 27:molecules27175458. [PMID: 36080226 PMCID: PMC9458238 DOI: 10.3390/molecules27175458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
Different aspects of intra-tumor heterogeneity (ITH), which are associated with the development of cancer and its response to treatment, have postulated prognostic value. Here we searched for potential association between phenotypic ITH analyzed by mass spectrometry imaging (MSI) and prognosis of head and neck cancer. The study involved tissue specimens resected from 77 patients with locally advanced oral squamous cell carcinoma, including 37 patients where matched samples of primary tumor and synchronous lymph node metastases were analyzed. A 3-year follow-up was available for all patients which enabled their separation into two groups: with no evidence of disease (NED, n = 41) and with progressive disease (PD, n = 36). After on-tissue trypsin digestion, peptide maps of all cancer regions were segmented using an unsupervised approach to reveal their intrinsic heterogeneity. We found that intra-tumor similarity of spectra was higher in the PD group and diversity of clusters identified during image segmentation was higher in the NED group, which indicated a higher level of ITH in patients with more favorable outcomes. Signature of molecular components that correlated with long-term outcomes could be associated with proteins involved in the immune functions. Furthermore, a positive correlation between ITH and histopathological lymphocytic host response was observed. Hence, we proposed that a higher level of ITH revealed by MSI in cancers with a better prognosis could reflect the presence of heterotypic components of tumor microenvironment such as infiltrating immune cells enhancing the response to the treatment.
Collapse
|
9
|
Janßen C, Boskamp T, Hauberg-Lotte L, Behrmann J, Deininger SO, Kriegsmann M, Kriegsmann K, Steinbuß G, Winter H, Muley T, Casadonte R, Kriegsmann J, Maaß P. Robust subtyping of non-small cell lung cancer whole sections through MALDI mass spectrometry imaging. Proteomics Clin Appl 2022; 16:e2100068. [PMID: 35238465 DOI: 10.1002/prca.202100068] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/24/2022] [Accepted: 02/28/2022] [Indexed: 12/30/2022]
Abstract
Subtyping of the most common non-small cell lung cancer (NSCLC) tumor types adenocarcinoma (ADC) and squamous cell carcinoma (SqCC) is still a challenge in the clinical routine and a correct diagnosis is crucial for an adequate therapy selection. Matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) has shown potential for NSCLC subtyping but is subject to strong technical variability and has only been applied to tissue samples assembled in tissue microarrays (TMAs). To our knowledge, a successful transfer of a classifier from TMAs to whole sections, which are generated in the standard clinical routine, has not been presented in the literature as of yet. We introduce a classification algorithm using extensive preprocessing and a classifier (either a neural network or a linear discriminant analysis (LDA)) to robustly classify whole sections of ADC and SqCC lung tissue. The classifiers were trained on TMAs and validated and tested on whole sections. Vital for a successful application on whole sections is the extensive preprocessing and the use of whole sections for hyperparameter selection. The classification system with the neural network/LDA results in 99.0%/98.3% test accuracy on spectra level and 100.0%/100.0% test accuracy on whole section level, respectively, and, therefore, provides a powerful tool to support the pathologist's decision making process. The presented method is a step further towards a clinical application of MALDI MSI and artificial intelligence for subtyping of NSCLC tissue sections.
Collapse
Affiliation(s)
- Charlotte Janßen
- Center for Industrial Mathematics (ZeTeM), University of Bremen, Bremen, Germany
| | - Tobias Boskamp
- Center for Industrial Mathematics (ZeTeM), University of Bremen, Bremen, Germany.,Bruker Daltonics GmbH, Bremen, Germany
| | - Lena Hauberg-Lotte
- Center for Industrial Mathematics (ZeTeM), University of Bremen, Bremen, Germany
| | - Jens Behrmann
- Center for Industrial Mathematics (ZeTeM), University of Bremen, Bremen, Germany
| | | | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Georg Steinbuß
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Surgery, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany.,Translational Research Unit, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Jörg Kriegsmann
- Proteopath, Trier, Germany.,Center for Histology, Cytology and Molecular Diagnostic, Trier, Germany
| | - Peter Maaß
- Center for Industrial Mathematics (ZeTeM), University of Bremen, Bremen, Germany
| |
Collapse
|
10
|
Aftab W, Lahiri S, Imhof A. ImShot: An Open-Source Software for Probabilistic Identification of Proteins In Situ and Visualization of Proteomics Data. Mol Cell Proteomics 2022; 21:100242. [PMID: 35569805 PMCID: PMC9194865 DOI: 10.1016/j.mcpro.2022.100242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/08/2022] [Accepted: 05/10/2022] [Indexed: 11/19/2022] Open
Abstract
Imaging mass spectrometry (IMS) has developed into a powerful tool allowing label-free detection of numerous biomolecules in situ. In contrast to shotgun proteomics, proteins/peptides can be detected directly from biological tissues and correlated to its morphology leading to a gain of crucial clinical information. However, direct identification of the detected molecules is currently challenging for MALDI-IMS, thereby compelling researchers to use complementary techniques and resource intensive experimental setups. Despite these strategies, sufficient information could not be extracted because of lack of an optimum data combination strategy/software. Here, we introduce a new open-source software ImShot that aims at identifying peptides obtained in MALDI-IMS. This is achieved by combining information from IMS and shotgun proteomics (LC-MS) measurements of serial sections of the same tissue. The software takes advantage of a two-group comparison to determine the search space of IMS masses after deisotoping the corresponding spectra. Ambiguity in annotations of IMS peptides is eliminated by introduction of a novel scoring system that identifies the most likely parent protein of a detected peptide in the corresponding IMS dataset. Thanks to its modular structure, the software can also handle LC-MS data separately and display interactive enrichment plots and enriched Gene Ontology terms or cellular pathways. The software has been built as a desktop application with a conveniently designed graphic user interface to provide users with a seamless experience in data analysis. ImShot can run on all the three major desktop operating systems and is freely available under Massachusetts Institute of Technology license.
Collapse
Affiliation(s)
- Wasim Aftab
- Biomedical Center, Protein Analysis Unit, Faculty of Medicine, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany; Graduate School for Quantitative Biosciences (QBM), Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Shibojyoti Lahiri
- Biomedical Center, Protein Analysis Unit, Faculty of Medicine, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| | - Axel Imhof
- Biomedical Center, Protein Analysis Unit, Faculty of Medicine, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
11
|
Applications of multivariate analysis and unsupervised machine learning to ToF-SIMS images of organic, bioorganic, and biological systems. Biointerphases 2022; 17:020802. [DOI: 10.1116/6.0001590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging offers a powerful, label-free method for exploring organic, bioorganic, and biological systems. The technique is capable of very high spatial resolution, while also producing an enormous amount of information about the chemical and molecular composition of a surface. However, this information is inherently complex, making interpretation and analysis of the vast amount of data produced by a single ToF-SIMS experiment a considerable challenge. Much research over the past few decades has focused on the application and development of multivariate analysis (MVA) and machine learning (ML) techniques that find meaningful patterns and relationships in these datasets. Here, we review the unsupervised algorithms—that is, algorithms that do not require ground truth labels—that have been applied to ToF-SIMS images, as well as other algorithms and approaches that have been used in the broader family of mass spectrometry imaging (MSI) techniques. We first give a nontechnical overview of several commonly used classes of unsupervised algorithms, such as matrix factorization, clustering, and nonlinear dimensionality reduction. We then review the application of unsupervised algorithms to various organic, bioorganic, and biological systems including cells and tissues, organic films, residues and coatings, and spatially structured systems such as polymer microarrays. We then cover several novel algorithms employed for other MSI techniques that have received little attention from ToF-SIMS imaging researchers. We conclude with a brief outline of potential future directions for the application of MVA and ML algorithms to ToF-SIMS images.
Collapse
|
12
|
Kelahan LC, Kim D, Soliman M, Avery RJ, Savas H, Agrawal R, Magnetta M, Liu BP, Velichko YS. Role of hepatic metastatic lesion size on inter-reader reproducibility of CT-based radiomics features. Eur Radiol 2022; 32:4025-4033. [PMID: 35080646 DOI: 10.1007/s00330-021-08526-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/05/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To evaluate the effect of hepatic metastatic lesion size on inter-reader reproducibility of CT-based 2D radiomics imaging features. METHODS Computerized tomography (CT) scans of 59 liver metastases from 34 patients with colorectal cancer were evaluated. Image segmentation was performed manually by three readers blinded to each other's results. For each radiomics feature, we created two datasets by sorting measurements according to size, i.e., (i) from the smallest to the largest lesion and (ii) from the largest to the smallest lesion. The Lin concordance correlation coefficient (CCC) was employed to analyze the reproducibility of radiomics features. In particular, the CCC was computed as a function of a number of elements in the dataset, by gradually adding lesions from each sorted dataset. To evaluate the effect of lesion size, we analyzed the difference between these two functions thus assessing the contribution of small and large lesions into the reproducibility of radiomics features. RESULTS Inter-reader reproducibility of CT-based 2D radiomics features assessed using Lin's CCC demonstrates tumor-size dependence. For example, the Lin CCC for GLCM contrast equals 0.88 (95% C.I. 0.84 to 0.92, p < 0.003) and could change by an additional + / - 0.06 depending on the presence of large or small lesions. CONCLUSIONS Groups of "large" and "small" lesions show different inter-reader reproducibility. The inter-reader reproducibility from the mixed group consisting of "large" and "small" lesions depends on the lesion-size distribution and can vary widely. This finding could partially explain variability in reproducibility of radiomics features in the literature. KEY POINTS • Groups of "large" and "small" lesions show different inter-reader reproducibility. • The inter-reader reproducibility from the mixed group consisting of "large" and "small" lesions depends on the lesion-size distribution.
Collapse
Affiliation(s)
- Linda C Kelahan
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Donald Kim
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Moataz Soliman
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Ryan J Avery
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Hatice Savas
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Rishi Agrawal
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Michael Magnetta
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Benjamin P Liu
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA
| | - Yuri S Velichko
- Department of Radiology, Northwestern University - Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL, 60611, USA.
| |
Collapse
|
13
|
Cordeiro YG, Mulder LM, van Zeijl RJM, Paskoski LB, van Veelen P, de Ru A, Strefezzi RF, Heijs B, Fukumasu H. Proteomic Analysis Identifies FNDC1, A1BG, and Antigen Processing Proteins Associated with Tumor Heterogeneity and Malignancy in a Canine Model of Breast Cancer. Cancers (Basel) 2021; 13:cancers13235901. [PMID: 34885011 PMCID: PMC8657005 DOI: 10.3390/cancers13235901] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/24/2022] Open
Abstract
New insights into the underlying biological processes of breast cancer are needed for the development of improved markers and treatments. The complex nature of mammary cancer in dogs makes it a great model to study cancer biology since they present a high degree of tumor heterogeneity. In search of disease-state biomarkers candidates, we applied proteomic mass spectrometry imaging in order to simultaneously detect histopathological and molecular alterations whilst preserving morphological integrity, comparing peptide expression between intratumor populations in distinct levels of differentiation. Peptides assigned to FNDC1, A1BG, and double-matching keratins 18 and 19 presented a higher intensity in poorly differentiated regions. In contrast, we observed a lower intensity of peptides matching calnexin, PDIA3, and HSPA5 in poorly differentiated cells, which enriched for protein folding in the endoplasmic reticulum and antigen processing, assembly, and loading of class I MHC. Over-representation of collagen metabolism, coagulation cascade, extracellular matrix components, cadherin-binding and cell adhesion pathways also distinguished cell populations. Finally, an independent validation showed FNDC1, A1BG, PDIA3, HSPA5, and calnexin as significant prognostic markers for human breast cancer patients. Thus, through a spatially correlated characterization of spontaneous carcinomas, we described key proteins which can be further validated as potential prognostic biomarkers.
Collapse
Affiliation(s)
- Yonara G. Cordeiro
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil; (Y.G.C.); (L.B.P.); (R.F.S.)
| | - Leandra M. Mulder
- Center of Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (L.M.M.); (R.J.M.v.Z.); (P.v.V.); (A.d.R.); (B.H.)
| | - René J. M. van Zeijl
- Center of Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (L.M.M.); (R.J.M.v.Z.); (P.v.V.); (A.d.R.); (B.H.)
| | - Lindsay B. Paskoski
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil; (Y.G.C.); (L.B.P.); (R.F.S.)
| | - Peter van Veelen
- Center of Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (L.M.M.); (R.J.M.v.Z.); (P.v.V.); (A.d.R.); (B.H.)
| | - Arnoud de Ru
- Center of Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (L.M.M.); (R.J.M.v.Z.); (P.v.V.); (A.d.R.); (B.H.)
| | - Ricardo F. Strefezzi
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil; (Y.G.C.); (L.B.P.); (R.F.S.)
| | - Bram Heijs
- Center of Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (L.M.M.); (R.J.M.v.Z.); (P.v.V.); (A.d.R.); (B.H.)
| | - Heidge Fukumasu
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil; (Y.G.C.); (L.B.P.); (R.F.S.)
- Correspondence: ; Tel.: +55-19-3565-6864
| |
Collapse
|
14
|
Gawin M, Kurczyk A, Niemiec J, Stanek-Widera A, Grela-Wojewoda A, Adamczyk A, Biskup-Frużyńska M, Polańska J, Widłak P. Intra-Tumor Heterogeneity Revealed by Mass Spectrometry Imaging Is Associated with the Prognosis of Breast Cancer. Cancers (Basel) 2021; 13:4349. [PMID: 34503159 PMCID: PMC8431441 DOI: 10.3390/cancers13174349] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Intra-tumor heterogeneity (ITH) results from the coexistence of genetically distinct cancer cell (sub)populations, their phenotypic plasticity, and the presence of heterotypic components of the tumor microenvironment (TME). Here we addressed the potential association between phenotypic ITH revealed by mass spectrometry imaging (MSI) and the prognosis of breast cancer. Tissue specimens resected from 59 patients treated radically due to the locally advanced HER2-positive invasive ductal carcinoma were included in the study. After the on-tissue trypsin digestion of cellular proteins, peptide maps of all cancer regions (about 380,000 spectra in total) were segmented by an unsupervised approach to reveal their intrinsic heterogeneity. A high degree of similarity between spectra was observed, which indicated the relative homogeneity of cancer regions. However, when the number and diversity of the detected clusters of spectra were analyzed, differences between patient groups were observed. It is noteworthy that a higher degree of heterogeneity was found in tumors from patients who remained disease-free during a 5-year follow-up (n = 38) compared to tumors from patients with progressive disease (distant metastases detected during the follow-up, n = 21). Interestingly, such differences were not observed between patients with a different status of regional lymph nodes, cancer grade, or expression of estrogen receptor at the time of the primary treatment. Subsequently, spectral components with different abundance in cancer regions were detected in patients with different outcomes, and their hypothetical identity was established by assignment to measured masses of tryptic peptides identified in corresponding tissue lysates. Such differentiating components were associated with proteins involved in immune regulation and hemostasis. Further, a positive correlation between the level of tumor-infiltrating lymphocytes and heterogeneity revealed by MSI was observed. We postulate that a higher heterogeneity of tumors with a better prognosis could reflect the presence of heterotypic components including infiltrating immune cells, that facilitated the response to treatment.
Collapse
Affiliation(s)
- Marta Gawin
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| | - Agata Kurczyk
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| | - Joanna Niemiec
- Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland; (J.N.); (A.G.-W.); (A.A.)
- Medical College of Rzeszow University, 35-959 Rzeszów, Poland
| | - Agata Stanek-Widera
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
- Faculty of Medicine, University of Technology in Katowice, 40-555 Katowice, Poland
| | - Aleksandra Grela-Wojewoda
- Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland; (J.N.); (A.G.-W.); (A.A.)
| | - Agnieszka Adamczyk
- Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland; (J.N.); (A.G.-W.); (A.A.)
| | - Magdalena Biskup-Frużyńska
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| | | | - Piotr Widłak
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.G.); (A.K.); (A.S.-W.); (M.B.-F.)
| |
Collapse
|
15
|
Dapic I, Baljeu-Neuman L, Uwugiaren N, Kers J, Goodlett DR, Corthals GL. Proteome analysis of tissues by mass spectrometry. MASS SPECTROMETRY REVIEWS 2019; 38:403-441. [PMID: 31390493 DOI: 10.1002/mas.21598] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/17/2019] [Indexed: 06/10/2023]
Abstract
Tissues and biofluids are important sources of information used for the detection of diseases and decisions on patient therapies. There are several accepted methods for preservation of tissues, among which the most popular are fresh-frozen and formalin-fixed paraffin embedded methods. Depending on the preservation method and the amount of sample available, various specific protocols are available for tissue processing for subsequent proteomic analysis. Protocols are tailored to answer various biological questions, and as such vary in lysis and digestion conditions, as well as duration. The existence of diverse tissue-sample protocols has led to confusion in how to choose the best protocol for a given tissue and made it difficult to compare results across sample types. Here, we summarize procedures used for tissue processing for subsequent bottom-up proteomic analysis. Furthermore, we compare protocols for their variations in the composition of lysis buffers, digestion procedures, and purification steps. For example, reports have shown that lysis buffer composition plays an important role in the profile of extracted proteins: the most common are tris(hydroxymethyl)aminomethane, radioimmunoprecipitation assay, and ammonium bicarbonate buffers. Although, trypsin is the most commonly used enzyme for proteolysis, in some protocols it is supplemented with Lys-C and/or chymotrypsin, which will often lead to an increase in proteome coverage. Data show that the selection of the lysis procedure might need to be tissue-specific to produce distinct protocols for individual tissue types. Finally, selection of the procedures is also influenced by the amount of sample available, which range from biopsies or the size of a few dozen of mm2 obtained with laser capture microdissection to much larger amounts that weight several milligrams.
Collapse
Affiliation(s)
- Irena Dapic
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
| | | | - Naomi Uwugiaren
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - David R Goodlett
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
- University of Maryland, 20N. Pine Street, Baltimore, MD 21201
| | - Garry L Corthals
- van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Kim KH, Ryu SY, Lee HY, Choi JY, Kwon OJ, Kim HK, Shim YM. Evaluating the tumor biology of lung adenocarcinoma: A multimodal analysis. Medicine (Baltimore) 2019; 98:e16313. [PMID: 31335678 PMCID: PMC6709045 DOI: 10.1097/md.0000000000016313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We evaluated the relationships among functional imaging modality such as PET-CT and DW-MRI and lung adenocarcinoma pathologic heterogeneity, extent of invasion depth, and tumor cellularity as a marker of tumor microenvironment.In total, 74 lung adenocarcinomas were prospectively included. All patients underwent 18F-fluorodeoxyglucose (FDG) PET-CT and MRI before curative surgery. Pathology revealed 68 stage I tumors, 3 stage II tumors, and 3 stage IIIA tumors. Comprehensive histologic subtyping was performed for all surgically resected tumors. Maximum standardized uptake value (SUVmax) and ADC values were correlated with pathologic grade, extent of invasion, solid tumor size, and tumor cellularity.Mean solid tumor size (low: 1.7 ± 3.0 mm, indeterminate: 13.9 ± 14.2 mm, and high grade: 30.3 ± 13.5 mm) and SUVmax (low: 1.5 ± 0.2, indeterminate: 3.5 ± 2.5, and high grade: 15.3 ± 0) had a significant relationship with pathologic grade based on 95% confidence intervals (P = .01 and P < .01, respectively). SUVmax showed a strong correlation with tumor cellularity (R = 0.713, P < .001), but was not correlated with extent of invasion (R = 0.387, P = .148). A significant and strong positive correlation was observed among SUVmax values and higher cellularity and pathologic grade. ADC did not exhibit a significant relationship with tumor cellularity.Intratumor heterogeneity quantification using a multimodal-multiparametric approach might be effective when tumor volume consists of a real tumor component as well as a non-tumorous stromal component.
Collapse
Affiliation(s)
- Ki Hwan Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
- Department of Radiology, Myongji Hospital, Goyang
| | - Seong-Yoon Ryu
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | | | - O. Jung Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Hong Kwan Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Young Mog Shim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Development and evaluation of matrix application techniques for high throughput mass spectrometry imaging of tissues in the clinic. CLINICAL MASS SPECTROMETRY 2019; 12:7-15. [DOI: 10.1016/j.clinms.2019.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 01/05/2023]
|
18
|
Restriction of drug transport by the tumor environment. Histochem Cell Biol 2018; 150:631-648. [DOI: 10.1007/s00418-018-1744-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2018] [Indexed: 12/31/2022]
|
19
|
Viewing the Future of IR through Molecular Histology: An Overview of Imaging Mass Spectrometry. J Vasc Interv Radiol 2018; 29:1543-1546.e1. [PMID: 30274858 DOI: 10.1016/j.jvir.2018.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/03/2018] [Accepted: 07/06/2018] [Indexed: 01/22/2023] Open
|
20
|
Tauler R, Parastar H. Big (Bio)Chemical Data Mining Using Chemometric Methods: A Need for Chemists. Angew Chem Int Ed Engl 2018; 61:e201801134. [DOI: 10.1002/anie.201801134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Roma Tauler
- IDAEA-CSIC Environmental Chemistry Jordi Girona 18-26 08034 Barcelona SPAIN
| | | |
Collapse
|
21
|
Zhai TT, van Dijk LV, Huang BT, Lin ZX, Ribeiro CO, Brouwer CL, Oosting SF, Halmos GB, Witjes MJH, Langendijk JA, Steenbakkers RJHM, Sijtsema NM. Improving the prediction of overall survival for head and neck cancer patients using image biomarkers in combination with clinical parameters. Radiother Oncol 2017; 124:256-262. [PMID: 28764926 DOI: 10.1016/j.radonc.2017.07.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/25/2017] [Accepted: 07/13/2017] [Indexed: 02/05/2023]
Abstract
PURPOSE To develop and validate prediction models of overall survival (OS) for head and neck cancer (HNC) patients based on image biomarkers (IBMs) of the primary tumor and positive lymph nodes (Ln) in combination with clinical parameters. MATERIAL AND METHODS The study cohort was composed of 289 nasopharyngeal cancer (NPC) patients from China and 298 HNC patients from the Netherlands. Multivariable Cox-regression analysis was performed to select clinical parameters from the NPC and HNC datasets, and IBMs from the NPC dataset. Final prediction models were based on both IBMs and clinical parameters. RESULTS Multivariable Cox-regression analysis identified three independent IBMs (tumor Volume-density, Run Length Non-uniformity and Ln Major-axis-length). This IBM model showed a concordance(c)-index of 0.72 (95%CI: 0.65-0.79) for the NPC dataset, which performed reasonably with a c-index of 0.67 (95%CI: 0.62-0.72) in the external validation HNC dataset. When IBMs were added in clinical models, the c-index of the NPC and HNC datasets improved to 0.75 (95%CI: 0.68-0.82; p=0.019) and 0.75 (95%CI: 0.70-0.81; p<0.001), respectively. CONCLUSION The addition of IBMs from the primary tumor and Ln improved the prognostic performance of the models containing clinical factors only. These combined models may improve pre-treatment individualized prediction of OS for HNC patients.
Collapse
Affiliation(s)
- Tian-Tian Zhai
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, China.
| | - Lisanne V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Bao-Tian Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, China
| | - Zhi-Xiong Lin
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, China.
| | - Cássia O Ribeiro
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Charlotte L Brouwer
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Sjoukje F Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Gyorgy B Halmos
- Department of Otolaryngology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Max J H Witjes
- Department of Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Johannes A Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Roel J H M Steenbakkers
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Nanna M Sijtsema
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| |
Collapse
|