1
|
Lee S, Arffman RK, Komsi EK, Lindgren O, Kemppainen J, Kask K, Saare M, Salumets A, Piltonen TT. Dynamic changes in AI-based analysis of endometrial cellular composition: Analysis of PCOS and RIF endometrium. J Pathol Inform 2024; 15:100364. [PMID: 38445292 PMCID: PMC10914580 DOI: 10.1016/j.jpi.2024.100364] [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: 12/15/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
Background The human endometrium undergoes a monthly cycle of tissue growth and degeneration. During the mid-secretory phase, the endometrium establishes an optimal niche for embryo implantation by regulating cellular composition (e.g., epithelial and stromal cells) and differentiation. Impaired endometrial development observed in conditions such as polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF) contributes to infertility. Surprisingly, despite the importance of the endometrial lining properly developing prior to pregnancy, precise measures of endometrial cellular composition in these two infertility-associated conditions are entirely lacking. Additionally, current methods for measuring the epithelial and stromal area have limitations, including intra- and inter-observer variability and efficiency. Methods We utilized a deep-learning artificial intelligence (AI) model, created on a cloud-based platform and developed in our previous study. The AI model underwent training to segment both areas populated by epithelial and stromal endometrial cells. During the training step, a total of 28.36 mm2 areas were annotated, comprising 2.56 mm2 of epithelium and 24.87 mm2 of stroma. Two experienced pathologists validated the performance of the AI model. 73 endometrial samples from healthy control women were included in the sample set to establish cycle phase-dependent dynamics of the endometrial epithelial-to-stroma ratio from the proliferative (PE) to secretory (SE) phases. In addition, 91 samples from PCOS cases, accounting for the presence or absence of ovulation and representing all menstrual cycle phases, and 29 samples from RIF patients on day 5 after progesterone administration in the hormone replacement treatment cycle were also included and analyzed in terms of cellular composition. Results Our AI model exhibited reliable and reproducible performance in delineating epithelial and stromal compartments, achieving an accuracy of 92.40% and 99.23%, respectively. Moreover, the performance of the AI model was comparable to the pathologists' assessment, with F1 scores exceeding 82% for the epithelium and >96% for the stroma. Next, we compared the endometrial epithelial-to-stromal ratio during the menstrual cycle in women with PCOS and in relation to endometrial receptivity status in RIF patients. The ovulatory PCOS endometrium exhibited epithelial cell proportions similar to those of control and healthy women's samples in every cycle phase, from the PE to the late SE, correlating with progesterone levels (control SE, r2 = 0.64, FDR < 0.001; PCOS SE, r2 = 0.52, FDR < 0.001). The mid-SE endometrium showed the highest epithelial percentage compared to both the early and late SE endometrium in both healthy women and PCOS patients. Anovulatory PCOS cases showed epithelial cellular fractions comparable to those of PCOS cases in the PE (Anovulatory, 14.54%; PCOS PE, 15.56%, p = 1.00). We did not observe significant differences in the epithelial-to-stroma ratio in the hormone-induced endometrium in RIF patients with different receptivity statuses. Conclusion The AI model rapidly and accurately identifies endometrial histology features by calculating areas occupied by epithelial and stromal cells. The AI model demonstrates changes in epithelial cellular proportions according to the menstrual cycle phase and reveals no changes in epithelial cellular proportions based on PCOS and RIF conditions. In conclusion, the AI model can potentially improve endometrial histology assessment by accelerating the analysis of the cellular composition of the tissue and by ensuring maximal objectivity for research and clinical purposes.
Collapse
Affiliation(s)
- Seungbaek Lee
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Riikka K. Arffman
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| | - Elina K. Komsi
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| | - Outi Lindgren
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Janette Kemppainen
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Keiu Kask
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm 14152, Sweden
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| |
Collapse
|
2
|
Lee S, Arffman RK, Komsi EK, Lindgren O, Kemppainen JA, Metsola H, Rossi HR, Ahtikoski A, Kask K, Saare M, Salumets A, Piltonen TT. AI-algorithm training and validation for identification of endometrial CD138+ cells in infertility-associated conditions; polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF). J Pathol Inform 2024; 15:100380. [PMID: 38827567 PMCID: PMC11140811 DOI: 10.1016/j.jpi.2024.100380] [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: 02/02/2024] [Revised: 04/20/2024] [Accepted: 04/26/2024] [Indexed: 06/04/2024] Open
Abstract
Background Endometrial CD138+ plasma cells serve as a diagnostic biomarker for endometrial inflammation, and their elevated occurrence correlates positively with adverse pregnancy outcomes. Infertility-related conditions like polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF) are closely associated with systemic and local chronic inflammatory status, wherein endometrial CD138+ plasma cell accumulation could also contribute to endometrial pathology. Current methods for quantifying CD138+ cells typically involve laborious and time-consuming microscopic assessments of only a few random areas from a slide. These methods have limitations in accurately representing the entire slide and are susceptible to significant biases arising from intra- and interobserver variations. Implementing artificial intelligence (AI) for CD138+ cell identification could enhance the accuracy, reproducibility, and reliability of analysis. Methods Here, an AI algorithm was developed to identify CD138+ plasma cells within endometrial tissue. The AI model comprised two layers of convolutional neural networks (CNNs). CNN1 was trained to segment epithelium and stroma across 28,363 mm2 (2.56 mm2 of epithelium and 24.87 mm2 of stroma), while CNN2 was trained to distinguish stromal cells based on CD138 staining, encompassing 7345 cells in the object layers (6942 CD138- cells and 403 CD138+ cells). The training and performance of the AI model were validated by three experienced pathologists. We collected 193 endometrial tissues from healthy controls (n = 73), women with PCOS (n = 91), and RIF patients (n = 29) and compared the CD138+ cell percentages based on cycle phases, ovulation status, and endometrial receptivity utilizing the AI model. Results The AI algorithm consistently and reliably distinguished CD138- and CD138+ cells, with total error rates of 6.32% and 3.23%, respectively. During the training validation, there was a complete agreement between the decisions made by the pathologists and the AI algorithm, while the performance validation demonstrated excellent accuracy between the AI and human evaluation methods (intraclass correlation; 0.76, 95% confidence intervals; 0.36-0.93, p = 0.002) and a positive correlation (Spearman's rank correlation coefficient: 0.79, p < 0.01). In the AI analysis, the AI model revealed higher CD138+ cell percentages in the proliferative phase (PE) endometrium compared to the secretory phase or anovulatory PCOS endometrium, irrespective of PCOS diagnosis. Interestingly, CD138+ percentages differed according to PCOS phenotype in the PE (p = 0.03). On the other hand, the receptivity status had no impact on the cell percentages in RIF samples. Conclusion Our findings emphasize the potential and accuracy of the AI algorithm in detecting endometrial CD138+ plasma cells, offering distinct advantages over manual inspection, such as rapid analysis of whole slide images, reduction of intra- and interobserver variations, sparing the valuable time of trained specialists, and consistent productivity. This supports the application of AI technology to help clinical decision-making, for example, in understanding endometrial cycle phase-related dynamics, as well as different reproductive disorders.
Collapse
Affiliation(s)
- Seungbaek Lee
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Riikka K. Arffman
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Elina K. Komsi
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Outi Lindgren
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Janette A. Kemppainen
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Hanna Metsola
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Henna-Riikka Rossi
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Anne Ahtikoski
- Department of Pathology, Turku University Hospital, Turku 20521, Finland
| | - Keiu Kask
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm 14152, Sweden
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| |
Collapse
|
3
|
Haga A, Iwasawa T, Misumi T, Okudela K, Oda T, Kitamura H, Saka T, Matsushita S, Baba T, Natsume-Kitatani Y, Utsunomiya D, Ogura T. Correlation of CT-based radiomics analysis with pathological cellular infiltration in fibrosing interstitial lung diseases. Jpn J Radiol 2024; 42:1157-1167. [PMID: 38888852 PMCID: PMC11442537 DOI: 10.1007/s11604-024-01607-2] [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: 02/08/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE We aimed to identify computed tomography (CT) radiomics features that are associated with cellular infiltration and construct CT radiomics models predictive of cellular infiltration in patients with fibrotic ILD. MATERIALS AND METHODS CT images of patients with ILD who underwent surgical lung biopsy (SLB) were analyzed. Radiomics features were extracted using artificial intelligence-based software and PyRadiomics. We constructed a model predicting cell counts in histological specimens, and another model predicting two classifications of higher or lower cellularity. We tested these models using external validation. RESULTS Overall, 100 patients (mean age: 62 ± 8.9 [standard deviation] years; 61 men) were included. The CT radiomics model used to predict cell count in 140 histological specimens predicted the actual cell count in 59 external validation specimens (root-mean-square error: 0.797). The two-classification model's accuracy was 70% and the F1 score was 0.73 in the external validation dataset including 30 patients. CONCLUSION The CT radiomics-based model developed in this study provided useful information regarding the cellular infiltration in the ILD with good correlation with SLB specimens.
Collapse
Affiliation(s)
- Akira Haga
- Dept. of Radiology, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan
- Dept. of Radiology, Yokohama City Univ. School of Medicine, Yokohama, Japan
| | - Tae Iwasawa
- Dept. of Radiology, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan.
| | - Toshihiro Misumi
- Department of Data Science, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koji Okudela
- Department of Pathology, Saitama Medical University, Moroyama, Japan
- Dept. of Pathology, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan
| | - Tsuneyuki Oda
- Dept. of Respiratory Medicine, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan
| | - Hideya Kitamura
- Dept. of Respiratory Medicine, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan
| | | | | | - Tomohisa Baba
- Dept. of Respiratory Medicine, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan
| | - Yayoi Natsume-Kitatani
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Daisuke Utsunomiya
- Dept. of Radiology, Yokohama City Univ. School of Medicine, Yokohama, Japan
| | - Takashi Ogura
- Dept. of Respiratory Medicine, Kanagawa Cardiovascular & Respiratory Center, Yokohama, Japan
| |
Collapse
|
4
|
Lavis P, Garabet A, Cardozo AK, Bondue B. The fibroblast activation protein alpha as a biomarker of pulmonary fibrosis. Front Med (Lausanne) 2024; 11:1393778. [PMID: 39364020 PMCID: PMC11446883 DOI: 10.3389/fmed.2024.1393778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rare, chronic, and progressive interstitial lung disease with an average survival of approximately 3 years. The evolution of IPF is unpredictable, with some patients presenting a relatively stable condition with limited progression over time, whereas others deteriorate rapidly. In addition to IPF, other interstitial lung diseases can lead to pulmonary fibrosis, and up to a third have a progressive phenotype with the same prognosis as IPF. Clinical, biological, and radiological risk factors of progression were identified, but no specific biomarkers of fibrogenesis are currently available. A recent interest in the fibroblast activation protein alpha (FAPα) has emerged. FAPα is a transmembrane serine protease with extracellular activity. It can also be found in a soluble form, also named anti-plasmin cleaving enzyme (APCE). FAPα is specifically expressed by activated fibroblasts, and quinoline-based specific inhibitors (FAPI) were developed, allowing us to visualize its distribution in vivo by imaging techniques. In this review, we discuss the use of FAPα as a useful biomarker for the progression of lung fibrosis, by both its assessment in human fluids and/or its detection by imaging techniques and immunohistochemistry.
Collapse
Affiliation(s)
- Philomène Lavis
- Department of Pathology, Hôpital universitaire de Bruxelles, Université libre de Bruxelles, Brussels, Belgium
- IRIBHM, Université libre de Bruxelles, Brussels, Belgium
| | - Ani Garabet
- Inflammation and Cell Death Signalling Group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Alessandra Kupper Cardozo
- Inflammation and Cell Death Signalling Group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Benjamin Bondue
- IRIBHM, Université libre de Bruxelles, Brussels, Belgium
- Department of Pneumology, Hôpital universitaire de Bruxelles, Université libre de Bruxelles, Brussels, Belgium
- European Reference Network for Rare Pulmonary Diseases (ERN-LUNG), Frankfurt, Germany
| |
Collapse
|
5
|
Li C, An Q, Jin Y, Jiang Z, Li M, Wu X, Dang H. Identification of oxidative stress-related diagnostic markers and immune infiltration features for idiopathic pulmonary fibrosis by bibliometrics and bioinformatics. Front Med (Lausanne) 2024; 11:1356825. [PMID: 39165378 PMCID: PMC11333355 DOI: 10.3389/fmed.2024.1356825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) garners considerable attention due to its high fatality rate and profound impact on quality of life. Our study conducts a comprehensive literature review on IPF using bibliometric analysis to explore existing hot research topics, and identifies novel diagnostic and therapeutic targets for IPF using bioinformatics analysis. Publications related to IPF from 2013 to 2023 were searched on the Web of Science Core Collection (WoSCC) database. Data analysis and visualization were conducted using CiteSpace and VOSviewer software primarily. The gene expression profiles GSE24206 and GSE53845 were employed as the training dataset. The GSE110147 dataset was employed as the validation dataset. We identified differentially expressed genes (DEGs) and differentially expressed genes related to oxidative stress (DEOSGs) between IPF and normal samples. Then, we conducted Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The hub genes were screened by protein-protein interaction (PPI) networks and machine learning algorithms. The CIBERSORT was used to analyze the immune infiltration of 22 kinds of immune cells. Finally, we conducted the expression and validation of hub genes. The diagnostic efficacy of hub genes was evaluated by employing Receiver Operating Characteristic (ROC) curves and the associations between hub genes and immune cells were analyzed. A total of 6,500 articles were identified, and the annual number of articles exhibited an upward trend. The United States emerged as the leading contributor in terms of publication count, institutional affiliations, highly cited articles, and prolific authorship. According to co-occurrence analysis, oxidative stress and inflammation are hot topics in IPF research. A total of 1,140 DEGs were identified, and 72 genes were classified as DEOSGs. By employing PPI network analysis and machine learning algorithms, PON2 and TLR4 were identified as hub genes. A total of 10 immune cells exhibited significant differences between IPF and normal samples. PON2 and TLR4, as oxidative stress-related genes, not only exhibit high diagnostic efficacy but also show close associations with immune cells. In summary, our study highlights oxidative stress and inflammation are hot topics in IPF research. Oxidative stress and immune cells play a vital role in the pathogenesis of IPF. Our findings suggest the potential of PON2 and TLR4 as novel diagnostic and therapeutic targets for IPF.
Collapse
Affiliation(s)
- Chang Li
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Qing An
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yi Jin
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zefei Jiang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meihe Li
- Department of Renal Transplantation, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoling Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huimin Dang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
6
|
John J, Clark AR, Kumar H, Burrowes KS, Vandal AC, Wilsher ML, Milne DG, Bartholmai BJ, Levin DL, Karwoski R, Tawhai MH. Evaluating Tissue Heterogeneity in the Radiologically Normal-Appearing Tissue in IPF Compared to Healthy Controls. Acad Radiol 2024; 31:1676-1685. [PMID: 37758587 DOI: 10.1016/j.acra.2023.08.046] [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: 07/20/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023]
Abstract
RATIONALE AND OBJECTIVES Idiopathic Pulmonary Fibrosis (IPF) is a progressive interstitial lung disease characterised by heterogeneously distributed fibrotic lesions. The inter- and intra-patient heterogeneity of the disease has meant that useful biomarkers of severity and progression have been elusive. Previous quantitative computed tomography (CT) based studies have focussed on characterising the pathological tissue. However, we hypothesised that the remaining lung tissue, which appears radiologically normal, may show important differences from controls in tissue characteristics. MATERIALS AND METHODS Quantitative metrics were derived from CT scans in IPF patients (N = 20) and healthy controls with a similar age (N = 59). An automated quantitative software (CALIPER, Computer-Aided Lung Informatics for Pathology Evaluation and Rating) was used to classify tissue as normal-appearing, fibrosis, or low attenuation area. Densitometry metrics were calculated for all lung tissue and for only the normal-appearing tissue. Heterogeneity of lung tissue density was quantified as coefficient of variation and by quadtree. Associations between measured lung function and quantitative metrics were assessed and compared between the two cohorts. RESULTS All metrics were significantly different between controls and IPF (p < 0.05), including when only the normal tissue was evaluated (p < 0.04). Density in the normal tissue was 14% higher in the IPF participants than controls (p < 0.001). The normal-appearing tissue in IPF had heterogeneity metrics that exhibited significant positive relationships with the percent predicted diffusion capacity for carbon monoxide. CONCLUSION We provide quantitative assessment of IPF lung tissue characteristics compared to a healthy control group of similar age. Tissue that appears visually normal in IPF exhibits subtle but quantifiable differences that are associated with lung function and gas exchange.
Collapse
Affiliation(s)
- Joyce John
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand (J.J., A.R.C., H.K., K.S.B., M.H.T.)
| | - Alys R Clark
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand (J.J., A.R.C., H.K., K.S.B., M.H.T.)
| | - Haribalan Kumar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand (J.J., A.R.C., H.K., K.S.B., M.H.T.)
| | - Kelly S Burrowes
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand (J.J., A.R.C., H.K., K.S.B., M.H.T.)
| | - Alain C Vandal
- Department of Statistics, University of Auckland, Auckland, New Zealand (A.C.V.)
| | - Margaret L Wilsher
- Respiratory Services, Auckland City Hospital, Auckland, New Zealand (M.L.W.)
| | - David G Milne
- Radiology, Auckland City Hospital, Auckland, New Zealand (D.G.M.)
| | | | - David L Levin
- Radiology, Mayo Clinic, Rochester, Minnesota (B.J.B., D.L.L., R.K.)
| | - Ronald Karwoski
- Radiology, Mayo Clinic, Rochester, Minnesota (B.J.B., D.L.L., R.K.)
| | - Merryn H Tawhai
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand (J.J., A.R.C., H.K., K.S.B., M.H.T.).
| |
Collapse
|
7
|
Lucà S, Pagliuca F, Perrotta F, Ronchi A, Mariniello DF, Natale G, Bianco A, Fiorelli A, Accardo M, Franco R. Multidisciplinary Approach to the Diagnosis of Idiopathic Interstitial Pneumonias: Focus on the Pathologist's Key Role. Int J Mol Sci 2024; 25:3618. [PMID: 38612431 PMCID: PMC11011777 DOI: 10.3390/ijms25073618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Idiopathic Interstitial Pneumonias (IIPs) are a heterogeneous group of the broader category of Interstitial Lung Diseases (ILDs), pathologically characterized by the distortion of lung parenchyma by interstitial inflammation and/or fibrosis. The American Thoracic Society (ATS)/European Respiratory Society (ERS) international multidisciplinary consensus classification of the IIPs was published in 2002 and then updated in 2013, with the authors emphasizing the need for a multidisciplinary approach to the diagnosis of IIPs. The histological evaluation of IIPs is challenging, and different types of IIPs are classically associated with specific histopathological patterns. However, morphological overlaps can be observed, and the same histopathological features can be seen in totally different clinical settings. Therefore, the pathologist's aim is to recognize the pathologic-morphologic pattern of disease in this clinical setting, and only after multi-disciplinary evaluation, if there is concordance between clinical and radiological findings, a definitive diagnosis of specific IIP can be established, allowing the optimal clinical-therapeutic management of the patient.
Collapse
Affiliation(s)
- Stefano Lucà
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.L.); (F.P.); (A.R.); (M.A.)
| | - Francesca Pagliuca
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.L.); (F.P.); (A.R.); (M.A.)
| | - Fabio Perrotta
- Department of Translational Medical Science, Università degli Studi della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (F.P.); (D.F.M.); (A.B.)
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.L.); (F.P.); (A.R.); (M.A.)
| | - Domenica Francesca Mariniello
- Department of Translational Medical Science, Università degli Studi della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (F.P.); (D.F.M.); (A.B.)
| | - Giovanni Natale
- Division of Thoracic Surgery, Università degli Studi della Campania “Luigi Vanvitelli”, Piazza Miraglia, 2, 80138 Naples, Italy; (G.N.); (A.F.)
| | - Andrea Bianco
- Department of Translational Medical Science, Università degli Studi della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (F.P.); (D.F.M.); (A.B.)
| | - Alfonso Fiorelli
- Division of Thoracic Surgery, Università degli Studi della Campania “Luigi Vanvitelli”, Piazza Miraglia, 2, 80138 Naples, Italy; (G.N.); (A.F.)
| | - Marina Accardo
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.L.); (F.P.); (A.R.); (M.A.)
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.L.); (F.P.); (A.R.); (M.A.)
| |
Collapse
|
8
|
Yi ES, Wawryko P, Ryu JH. Diagnosis of interstitial lung diseases: from Averill A. Liebow to artificial intelligence. J Pathol Transl Med 2024; 58:1-11. [PMID: 38229429 DOI: 10.4132/jptm.2023.11.17] [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: 10/23/2023] [Accepted: 11/17/2023] [Indexed: 01/18/2024] Open
Abstract
Histopathologic criteria of usual interstitial pneumonia (UIP)/idiopathic pulmonary fibrosis (IPF) were defined over the years and endorsed by leading organizations decades after Dr. Averill A. Liebow first coined the term UIP in the 1960s as a distinct pathologic pattern of fibrotic interstitial lung disease. Novel technology and recent research on interstitial lung diseases with genetic component shed light on molecular pathogenesis of UIP/IPF. Two antifibrotic agents introduced in the mid-2010s opened a new era of therapeutic approaches to UIP/IPF, albeit contentious issues regarding their efficacy, side effects, and costs. Recently, the concept of progressive pulmonary fibrosis was introduced to acknowledge additional types of progressive fibrosing interstitial lung diseases with the clinical and pathologic phenotypes comparable to those of UIP/IPF. Likewise, some authors have proposed a paradigm shift by considering UIP as a stand-alone diagnostic entity to encompass other fibrosing interstitial lung diseases that manifest a relentless progression as in IPF. These trends signal a pendulum moving toward the tendency of lumping diagnoses, which poses a risk of obscuring potentially important information crucial to both clinical and research purposes. Recent advances in whole slide imaging for digital pathology and artificial intelligence technology could offer an unprecedented opportunity to enhance histopathologic evaluation of interstitial lung diseases. However, current clinical practice trends of moving away from surgical lung biopsies in interstitial lung disease patients may become a limiting factor in this endeavor as it would be difficult to build a large histopathologic database with correlative clinical data required for artificial intelligence models.
Collapse
Affiliation(s)
- Eunhee S Yi
- Division of Anatomic Pathology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Paul Wawryko
- Division of Anatomic Pathology, Mayo Clinic Arizona, Arizona, FL, USA
| | - Jay H Ryu
- Division of Pulmonary and Critical Medicine, Mayo Clinic Rochester, Rochester, MN, USA
| |
Collapse
|
9
|
Handa T. The potential role of artificial intelligence in the clinical practice of interstitial lung disease. Respir Investig 2023; 61:702-710. [PMID: 37708636 DOI: 10.1016/j.resinv.2023.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/26/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023]
Abstract
Artificial intelligence (AI) is being widely applied in the field of medicine, in areas such as drug discovery, diagnostic support, and assistance with medical practice. Among these, medical imaging is an area where AI is expected to make a significant contribution. In Japan, as of November 2022, 23 AI medical devices have received regulatory approval; all these devices are related to image analysis. In interstitial lung diseases, technologies have been developed that use AI to analyze high-resolution computed tomography and pathological images, and gene expression patterns in tissue taken from transbronchial lung biopsies to assist in the diagnosis of idiopathic pulmonary fibrosis. Some of these technologies are already being used in clinical practice in the United States. AI is expected to reduce the burden on physicians, improve reproducibility, and advance personalized medicine. Obtaining sufficient data for diseases with a small number of patients is difficult. Additionally, certain issues must be addressed in order for AI to be applied in healthcare. These issues include taking responsibility for the AI results output, updating software after the launch of technology, and adapting to new imaging technologies. Establishing research infrastructures such as large-scale databases and common platforms is important for the development of AI technology: their use requires an understanding of the characteristics and limitations of the systems. CLINICAL TRIAL REGISTRATION: Not applicable.
Collapse
Affiliation(s)
- Tomohiro Handa
- Department of Advanced Medicine for Respiratory Failure and Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
10
|
Yang S, Wang J, Sun D, Wang Y, Xue C, Ye Q. Disease progression in patients with usual interstitial pneumonia and probable UIP patterns on computed tomography with various underlying etiologies: a retrospective cohort study. Front Med (Lausanne) 2023; 10:1246767. [PMID: 37901393 PMCID: PMC10601466 DOI: 10.3389/fmed.2023.1246767] [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: 06/24/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
Background Usual interstitial pneumonia (UIP) is a pattern of interstitial pneumonia that is caused by different etiologies. This study aimed to investigate the transplant-free survival (TFS) and the decline in forced vital capacity (FVC) of the patients with UIP and probable UIP patterns on CT caused by various underlying conditions. Methods A retrospective cohort study was conducted, enrolling patients with interstitial lung disease exhibiting a CT pattern consistent with UIP or probable UIP. Clinical and prognostic data of patients categorized by the etiology were compared. Results A total of 591 patients were included and classified into the following groups: idiopathic pulmonary fibrosis (IPF) (n = 320), connective tissue disease (CTD)-UIP (n = 229), asbestosis-UIP (n = 28), and hypersensitivity pneumonitis (HP)-UIP (n = 14). Advanced age, elevated levels of serum cytokeratin fraction 21-1 and percentage of neutrophils in bronchoalveolar lavage were observed in all groups. IPF patients showed a more rapid decline in FVC (133.9 mL/year) compared to CTD-UIP (24.5 mL/year, p = 0.001) and asbestosis-UIP (61.0 mL/year, p = 0.008) respectively. Sub-analysis of CTD-UIP revealed that patients with rheumatoid arthritis (RA)-UIP (88.1 mL/year) or antineutrophil cytoplasmic antibody-associated vasculitis (AAV)-UIP (72.9 mL/year) experienced a faster deterioration in FVC compared to those with primary Sjögren's syndrome (pSS)-UIP (25.9 mL/year, p < 0.05). Kaplan-Meier curves showed that IPF had the poorest TFS (median 55.9 months), followed by HP-UIP (57.5 months), CTD-UIP (66.7 months), and asbestosis-UIP (TFS not reached). RA-UIP or AAV-UIP did not exhibit any prognostic advantages compared to IPF, while asbestosis-UIP and pSS-UIP showed better survival rates. Conclusion Patients with UIP caused by different underlying conditions share certain common features, but the trajectories of disease progression and survival outcomes differ.
Collapse
Affiliation(s)
- Shuqiao Yang
- Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jing Wang
- Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Di Sun
- Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Occupational Medicine and Toxicology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yiran Wang
- Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Occupational Medicine and Toxicology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changjiang Xue
- Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Occupational Medicine and Toxicology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qiao Ye
- Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Occupational Medicine and Toxicology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Lew D, Klang E, Soffer S, Morgenthau AS. Current Applications of Artificial Intelligence in Sarcoidosis. Lung 2023; 201:445-454. [PMID: 37730926 DOI: 10.1007/s00408-023-00641-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/15/2023] [Indexed: 09/22/2023]
Abstract
PURPOSE Sarcoidosis is a complex disease which can affect nearly every organ system with manifestations ranging from asymptomatic imaging findings to sudden cardiac death. As such, diagnosis and prognostication are topics of continued investigation. Recent technological advancements have introduced multiple modalities of artificial intelligence (AI) to the study of sarcoidosis. Machine learning, deep learning, and radiomics have predominantly been used to study sarcoidosis. METHODS Articles were collected by searching online databases using keywords such as sarcoid, machine learning, artificial intelligence, radiomics, and deep learning. Article titles and abstracts were reviewed for relevance by a single reviewer. Articles written in languages other than English were excluded. CONCLUSIONS Machine learning may be used to help diagnose pulmonary sarcoidosis and prognosticate in cardiac sarcoidosis. Deep learning is most comprehensively studied for diagnosis of pulmonary sarcoidosis and has less frequently been applied to prognostication in cardiac sarcoidosis. Radiomics has primarily been used to differentiate sarcoidosis from malignancy. To date, the use of AI in sarcoidosis is limited by the rarity of this disease, leading to small, suboptimal training sets. Nevertheless, there are applications of AI that have been used to study other systemic diseases, which may be adapted for use in sarcoidosis. These applications include discovery of new disease phenotypes, discovery of biomarkers of disease onset and activity, and treatment optimization.
Collapse
Affiliation(s)
- Dana Lew
- Division of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eyal Klang
- Department of Diagnostic Imaging, Sheba Medical Center, Ramat Gan, Israel
| | - Shelly Soffer
- Division of Internal Medicine, Assuta Medical Center, Ashdod, Israel
| | - Adam S Morgenthau
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Mancini M, Bargiacchi L, De Vitis C, D'Ascanio M, De Dominicis C, Ibrahim M, Rendina EA, Ricci A, Di Napoli A, Mancini R, Vecchione A. Histologic Analysis of Idiopathic Pulmonary Fibrosis by Morphometric and Fractal Analysis. Biomedicines 2023; 11:biomedicines11051483. [PMID: 37239155 DOI: 10.3390/biomedicines11051483] [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: 04/04/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrotic lung disorder, ultimately leading to respiratory failure and death. Despite great research advances in understanding the mechanisms underlying the disease, its diagnosis, and its treatment, IPF still remains idiopathic without known biological or histological markers able to predict disease progression or response to treatment. The histologic hallmark of IPF is usual interstitial pneumonia (UIP), with its intricate architectural distortion and temporal inhomogeneity. We hypothesize that normal lung alveolar architecture can be compared to fractals, such as the Pythagoras tree with its fractal dimension (Df), and every pathological insult, distorting the normal lung structure, could result in Df variations. In this study, we aimed to assess the UIP histologic fractal dimension in relationship to other morphometric parameters in newly diagnosed IPF patients and its possible role in the prognostic stratification of the disease. Clinical data and lung tissue specimens were obtained from twelve patients with IPF, twelve patients with non-specific interstitial pneumonia (NSIP), and age-matched "healthy" control lung tissue from patients undergoing lung surgery for other causes. Histology and histomorphometry were performed to evaluate Df and lacunarity measures, using the box counting method on the FracLac ImageJ plugin. The results showed that Df was significantly higher in IPF patients compared to controls and fibrotic NSIP patients, indicating greater architectural distortion in IPF. Additionally, high Df values were associated with higher fibroblastic foci density and worse prognostic outcomes in IPF, suggesting that Df may serve as a potential novel prognostic marker for IPF. The scalability of Df measurements was demonstrated through repeated measurements on smaller portions from the same surgical biopsies, which were selected to mimic a cryobiopsy. Our study provides further evidence to support the use of fractal morphometry as a tool for quantifying and determining lung tissue remodeling in IPF, and we demonstrated a significant correlation between histological and radiological Df in UIP pattern, as well as a significant association between Df and FF density. Furthermore, our study demonstrates the scalability and self-similarity of Df measurements across different biopsy types, including surgical and smaller specimens.
Collapse
Affiliation(s)
- Massimiliano Mancini
- Morphologic and Molecular Pathology Unit, Sant'Andrea University Hospital, 00189 Rome, Italy
| | - Lavinia Bargiacchi
- Morphologic and Molecular Pathology Unit, Sant'Andrea University Hospital, 00189 Rome, Italy
| | - Claudia De Vitis
- Department of Clinical and Molecular Medicine Sant'Andrea University Hospital, "Sapienza" University of Rome", 00189 Rome, Italy
| | - Michela D'Ascanio
- UOC Respiratory Disease, Sant'Andrea University Hospital, 00189 Rome, Italy
| | | | - Mohsen Ibrahim
- Thoracic Surgery Unit, Sant'Andrea University Hospital, "Sapienza" University of Rome, 00189 Rome, Italy
| | - Erino Angelo Rendina
- Thoracic Surgery Unit, Sant'Andrea University Hospital, "Sapienza" University of Rome, 00189 Rome, Italy
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine Sant'Andrea University Hospital, "Sapienza" University of Rome", 00189 Rome, Italy
| | - Arianna Di Napoli
- Department of Clinical and Molecular Medicine Sant'Andrea University Hospital, "Sapienza" University of Rome", 00189 Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine Sant'Andrea University Hospital, "Sapienza" University of Rome", 00189 Rome, Italy
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine Sant'Andrea University Hospital, "Sapienza" University of Rome", 00189 Rome, Italy
| |
Collapse
|
13
|
Li Z, Wang S, Zhao H, Yan P, Yuan H, Zhao M, Wan R, Yu G, Wang L. Artificial neural network identified the significant genes to distinguish Idiopathic pulmonary fibrosis. Sci Rep 2023; 13:1225. [PMID: 36681777 PMCID: PMC9867697 DOI: 10.1038/s41598-023-28536-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease that causes irreversible damage to lung tissue characterized by excessive deposition of extracellular matrix (ECM) and remodeling of lung parenchyma. The current diagnosis of IPF is complex and usually completed by a multidisciplinary team including clinicians, radiologists and pathologists they work together and make decision for an effective treatment, it is imperative to introduce novel practical methods for IPF diagnosis. This study provided a new diagnostic model of idiopathic pulmonary fibrosis based on machine learning. Six genes including CDH3, DIO2, ADAMTS14, HS6ST2, IL13RA2, and IGFL2 were identified based on the differentially expressed genes in IPF patients compare to healthy subjects through a random forest classifier with the existing gene expression databases. An artificial neural network model was constructed for IPF diagnosis based these genes, and this model was validated by the distinctive public datasets with a satisfactory diagnostic accuracy. These six genes identified were significant correlated with lung function, and among them, CDH3 and DIO2 were further determined to be significantly associated with the survival. Putting together, artificial neural network model identified the significant genes to distinguish idiopathic pulmonary fibrosis from healthy people and it is potential for molecular diagnosis of IPF.
Collapse
Affiliation(s)
- Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Shenghui Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Huabin Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Peishuo Yan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Hongmei Yuan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Mengxia Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Ruyan Wan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China.
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 453007, Henan, China.
| |
Collapse
|
14
|
Glenn LM, Troy LK, Corte TJ. Novel diagnostic techniques in interstitial lung disease. Front Med (Lausanne) 2023; 10:1174443. [PMID: 37188089 PMCID: PMC10175799 DOI: 10.3389/fmed.2023.1174443] [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: 02/26/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Research into novel diagnostic techniques and targeted therapeutics in interstitial lung disease (ILD) is moving the field toward increased precision and improved patient outcomes. An array of molecular techniques, machine learning approaches and other innovative methods including electronic nose technology and endobronchial optical coherence tomography are promising tools with potential to increase diagnostic accuracy. This review provides a comprehensive overview of the current evidence regarding evolving diagnostic methods in ILD and to consider their future role in routine clinical care.
Collapse
Affiliation(s)
- Laura M. Glenn
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Central Clinical School, The University of Sydney School of Medicine, Sydney, NSW, Australia
- NHMRC Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
- *Correspondence: Laura M. Glenn,
| | - Lauren K. Troy
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Central Clinical School, The University of Sydney School of Medicine, Sydney, NSW, Australia
- NHMRC Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
| | - Tamera J. Corte
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Central Clinical School, The University of Sydney School of Medicine, Sydney, NSW, Australia
- NHMRC Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
| |
Collapse
|
15
|
Masanam HB, Perumal G, Krishnan S, Singh SK, Jha NK, Chellappan DK, Dua K, Gupta PK, Narasimhan AK. Advances and opportunities in nanoimaging agents for the diagnosis of inflammatory lung diseases. Nanomedicine (Lond) 2022; 17:1981-2005. [PMID: 36695290 DOI: 10.2217/nnm-2021-0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The development of rapid, noninvasive diagnostics to detect lung diseases is a great need after the COVID-2019 outbreak. The nanotechnology-based approach has improved imaging and facilitates the early diagnosis of inflammatory lung diseases. The multifunctional properties of nanoprobes enable better spatial-temporal resolution and a high signal-to-noise ratio in imaging. Targeted nanoimaging agents have been used to bind specific tissues in inflammatory lungs for early-stage diagnosis. However, nanobased imaging approaches for inflammatory lung diseases are still in their infancy. This review provides a solution-focused approach to exploring medical imaging technologies and nanoprobes for the detection of inflammatory lung diseases. Prospects for the development of contrast agents for lung disease detection are also discussed.
Collapse
Affiliation(s)
- Hema Brindha Masanam
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - Govindaraj Perumal
- Department of Conservative Dentistry & Endodontics, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Velappanchavadi, Chennai, 600 077, India.,Department of Biomedical Engineering, Rajalakshmi Engineering College, Thandalam, Chennai, 602 105, India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences & Research (SBSR), Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India.,Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India.,Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Ashwin Kumar Narasimhan
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, 603 203, India
| |
Collapse
|
16
|
Stetzik L, Mercado G, Smith L, George S, Quansah E, Luda K, Schulz E, Meyerdirk L, Lindquist A, Bergsma A, Jones RG, Brundin L, Henderson MX, Pospisilik JA, Brundin P. A novel automated morphological analysis of Iba1+ microglia using a deep learning assisted model. Front Cell Neurosci 2022; 16:944875. [PMID: 36187297 PMCID: PMC9520629 DOI: 10.3389/fncel.2022.944875] [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: 05/16/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023] Open
Abstract
There is growing evidence for the key role of microglial functional state in brain pathophysiology. Consequently, there is a need for efficient automated methods to measure the morphological changes distinctive of microglia functional states in research settings. Currently, many commonly used automated methods can be subject to sample representation bias, time consuming imaging, specific hardware requirements and difficulty in maintaining an accurate comparison across research environments. To overcome these issues, we use commercially available deep learning tools Aiforia® Cloud (Aifoira Inc., Cambridge, MA, United States) to quantify microglial morphology and cell counts from histopathological slides of Iba1 stained tissue sections. We provide evidence for the effective application of this method across a range of independently collected datasets in mouse models of viral infection and Parkinson's disease. Additionally, we provide a comprehensive workflow with training details and annotation strategies by feature layer that can be used as a guide to generate new models. In addition, all models described in this work are available within the Aiforia® platform for study-specific adaptation and validation.
Collapse
Affiliation(s)
- Lucas Stetzik
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States,*Correspondence: Lucas Stetzik,
| | - Gabriela Mercado
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Lindsey Smith
- Aiforia Inc, Cambridge Innovation Center, Cambridge, MA, United States
| | - Sonia George
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Emmanuel Quansah
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Katarzyna Luda
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, United States
| | - Emily Schulz
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Lindsay Meyerdirk
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Allison Lindquist
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Alexis Bergsma
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, United States
| | - Russell G. Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, United States
| | - Lena Brundin
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Michael X. Henderson
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | | | - Patrik Brundin
- Parkinson’s Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| |
Collapse
|
17
|
Talbott HE, Mascharak S, Griffin M, Wan DC, Longaker MT. Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell 2022; 29:1161-1180. [PMID: 35931028 PMCID: PMC9357250 DOI: 10.1016/j.stem.2022.07.006] [Citation(s) in RCA: 189] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibroblasts are highly dynamic cells that play a central role in tissue repair and fibrosis. However, the mechanisms by which they contribute to both physiologic and pathologic states of extracellular matrix deposition and remodeling are just starting to be understood. In this review article, we discuss the current state of knowledge in fibroblast biology and heterogeneity, with a primary focus on the role of fibroblasts in skin wound repair. We also consider emerging techniques in the field, which enable an increasingly nuanced and contextualized understanding of these complex systems, and evaluate limitations of existing methodologies and knowledge. Collectively, this review spotlights a diverse body of research examining an often-overlooked cell type-the fibroblast-and its critical functions in wound repair and beyond.
Collapse
Affiliation(s)
- Heather E Talbott
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
19
|
Usual interstitial pneumonia: a clinically significant pattern, but not the final word. Mod Pathol 2022; 35:589-593. [PMID: 35210554 DOI: 10.1038/s41379-022-01054-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/08/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022]
Abstract
Usual interstitial pneumonia (UIP) is a concept that is deeply entrenched in clinical practice and the prognostic significance of UIP is well established, but the field continues to suffer from the lack of a true gold standard for diagnosing fibrotic interstitial lung disease (ILD). The meaning and usage of UIP have shifted over time and this term is prone to misinterpretation and poor diagnostic agreement. For pathologists, it is worth reflecting on the limitations of UIP and our true role in the care of patients with ILD, a controversial topic explored in two point-counterpoint editorials published simultaneously in this journal. Current diagnostic guidelines are ambiguous and difficult to apply in clinical practice. Further complicating matters for the pathologist is the paradigm shift that occurred with the advent of anti-fibrotic agents, necessitating increased focus on the most likely etiology of fibrosis rather than simply the pattern of fibrosis when pulmonologists select appropriate therapy. Despite the wealth of information locked in tissue samples that could provide novel insights into fibrotic ILDs, pulmonologists increasingly shy away from obtaining biopsies, likely because pathologists no longer provide sufficient value to offset the risks of a biopsy procedure, and pathologic assessment is insufficiently reliable to meaningfully inform therapeutic decisionmaking. To increase the value of biopsies, pathologists must first recognize the problems with UIP as a diagnostic term. Second, pathologists must realize that the primary goal of a biopsy is to determine the most likely etiology to target with therapy, requiring a shift in diagnostic focus. Third, pathologists must devise and validate new classifications and criteria that are evidence-based, biologically relevant, easy to use, and predictive of outcome and treatment response. Only after the limitations of UIP are understood will pathologists provide maximum diagnostic value from biopsies to clinicians today and advance the field forward.
Collapse
|
20
|
Álvarez-Vásquez JL, Castañeda-Alvarado CP. Dental pulp fibroblast: A star Cell. J Endod 2022; 48:1005-1019. [DOI: 10.1016/j.joen.2022.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/16/2022]
|
21
|
Balancin ML, Baldavira CM, Prieto TG, Machado-Rugolo J, Farhat C, Assato AK, Velosa APP, Teodoro WR, Ab'Saber AM, Takagaki TY, Capelozzi VL. Dissecting and Reconstructing Matrix in Malignant Mesothelioma Through Histocell-Histochemistry Gradients for Clinical Applications. Front Med (Lausanne) 2022; 9:871202. [PMID: 35492318 PMCID: PMC9043486 DOI: 10.3389/fmed.2022.871202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMalignant pleural mesotheliomas (MM) are known for their heterogenous histology and clinical behavior. MM histology reveals three major tumor cell populations: epithelioid, sarcomatoid, and biphasic. Using a dissecting approach, we showed that histochemical gradients help us better understand tumor heterogeneity and reconsider its histologic classifications. We also showed that this method to characterize MM tumor cell populations provides a better understanding of the underlying mechanisms for invasion and disease progression.MethodsIn a cohort of 87 patients with surgically excised MM, we used hematoxylin and eosin to characterize tumor cell populations and Movat's pentachrome staining to dissect the ECM matrisome. Next, we developed a computerized semi-assisted protocol to quantify and reconstruct the ECM in 3D and examined the clinical association between the matricellular factors and patient outcome.ResultsEpithelioid cells had a higher matrix composition of elastin and fibrin, whereas, in the sarcomatoid type, hyaluronic acid and total collagen were most prevalent. The 3D reconstruction exposed the collagen I and III that form channels surrounding the neoplastic cell blocks. The estimated volume of the two collagen fractions was 14% of the total volume, consistent with the median estimated area of total collagen (12.05 mm2) for epithelioid MM.ConclusionDifferential patterns in matricellular phenotypes in MM could be used in translational studies to improve patient outcome. More importantly, our data raise the possibility that cancer cells can use the matrisome for disease expansion and could be effectively targeted by anti-collagen, anti-elastin, and/or anti-hyaluronic acid therapies.
Collapse
Affiliation(s)
- Marcelo Luiz Balancin
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Camila Machado Baldavira
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Tabatha Gutierrez Prieto
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Juliana Machado-Rugolo
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
- Health Technology Assessment Center (NATS), Clinical Hospital (HCFMB), Medical School of São Paulo State University (UNESP), Botucatu, Brazil
| | - Cecília Farhat
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Aline Kawassaki Assato
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, FMUSP, São Paulo, Brazil
| | - Walcy Rosolia Teodoro
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, FMUSP, São Paulo, Brazil
| | - Alexandre Muxfeldt Ab'Saber
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Teresa Yae Takagaki
- Division of Pneumology, Instituto do Coração (Incor), University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
- *Correspondence: Vera Luiza Capelozzi
| |
Collapse
|
22
|
RNA Sequencing of Epithelial Cell/Fibroblastic Foci Sandwich in Idiopathic Pulmonary Fibrosis: New Insights on the Signaling Pathway. Int J Mol Sci 2022; 23:ijms23063323. [PMID: 35328744 PMCID: PMC8954546 DOI: 10.3390/ijms23063323] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease characterized by irreversible scarring of the distal lung. IPF is best described by its histopathological pattern of usual interstitial pneumonia (UIP), characterized by spatial heterogeneity with alternating interstitial fibrosis and areas of normal lung, and temporal heterogeneity of fibrosis characterized by scattered fibroblastic foci (FF), dense acellular collagen and honeycomb changes. FF, comprising aggregated fibroblasts/myofibroblasts surrounded by metaplastic epithelial cells (EC), are the cardinal pathological lesion and their presence strongly correlates with disease progression and mortality. We hypothesized that the EC/FF sandwich from patients with UIP/IPF has a distinct molecular signature which could offer new insights into the crosstalk of these two crucial actors in the disease. Laser capture microdissection with RNAseq was used to investigate the transcriptome of the EC/FF sandwich from IPF patients versus controls (primary spontaneous pneumothorax). Differentially expressed gene analysis identified 23 up-regulated genes mainly related to epithelial dysfunction. Gene ontology analysis highlighted the activation of different pathways, mainly related to EC, immune response and programmed cell death. This study provides novel insights into the IPF pathogenetic pathways and suggests that targeting some of these up-regulated pathways (particularly those related to secreto-protein/mucin dysfunction) may be beneficial in IPF. Further studies in a larger number of lung samples, ideally from patients with early and advanced disease, are needed to validate these findings.
Collapse
|
23
|
Cau R, Faa G, Nardi V, Balestrieri A, Puig J, Suri JS, SanFilippo R, Saba L. Long-COVID diagnosis: From diagnostic to advanced AI-driven models. Eur J Radiol 2022; 148:110164. [PMID: 35114535 PMCID: PMC8791239 DOI: 10.1016/j.ejrad.2022.110164] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
Abstract
SARS-COV 2 is recognized to be responsible for a multi-organ syndrome. In most patients, symptoms are mild. However, in certain subjects, COVID-19 tends to progress more severely. Most of the patients infected with SARS-COV2 fully recovered within some weeks. In a considerable number of patients, like many other viral infections, various long-lasting symptoms have been described, now defined as "long COVID-19 syndrome". Given the high number of contagious over the world, it is necessary to understand and comprehend this emerging pathology to enable early diagnosis and improve patents outcomes. In this scenario, AI-based models can be applied in long-COVID-19 patients to assist clinicians and at the same time, to reduce the considerable impact on the care and rehabilitation unit. The purpose of this manuscript is to review different aspects of long-COVID-19 syndrome from clinical presentation to diagnosis, highlighting the considerable impact that AI can have.
Collapse
Affiliation(s)
- Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari - Polo di Monserrato, s.s. 554 Monserrato, Cagliari 09045, Italy
| | - Gavino Faa
- Department of Pathology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari, Italy
| | - Valentina Nardi
- Department of Cardiovascular Medicine Mayo Clinic, Rochester, MN, USA
| | - Antonella Balestrieri
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari - Polo di Monserrato, s.s. 554 Monserrato, Cagliari 09045, Italy
| | - Josep Puig
- Department of Radiology (IDI), Hospital Universitari de Girona, Girona, Spain
| | - Jasjit S Suri
- Stroke Diagnosis and Monitoring Division, Atheropoint LLC, Roseville, CA, USA
| | - Roberto SanFilippo
- Department of Vascular Surgery, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari - Polo di Monserrato, s.s. 554 Monserrato, Cagliari 09045, Italy
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari - Polo di Monserrato, s.s. 554 Monserrato, Cagliari 09045, Italy.
| |
Collapse
|
24
|
The histologic diagnosis of usual interstitial pneumonia of idiopathic pulmonary fibrosis. Where we are and where we need to go. Mod Pathol 2022; 35:8-14. [PMID: 34465882 PMCID: PMC8695374 DOI: 10.1038/s41379-021-00889-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/19/2021] [Accepted: 07/31/2021] [Indexed: 11/08/2022]
Abstract
In the 50 years since its inception by Dr. Liebow, the diagnosis of usual interstitial pneumonia (UIP) by pathologists has changed significantly. This manuscript reviews the progressive history of the histologic diagnosis of UIP and summarizes the current state of histologic UIP and its relationship to the clinical syndrome idiopathic pulmonary fibrosis (IPF). Fibrotic lung disease mimics of UIP/IPF are reviewed and pearls for distinguishing these diseases from UIP/IPF are provided. Strategies for increasing the value of histologic assessment of biopsies in the setting of pulmonary fibrosis are also discussed.
Collapse
|
25
|
van Batenburg AA, van Oosterhout MFM, Knoppert SN, Kazemier KM, van der Vis JJ, Grutters JC, Goldschmeding R, van Moorsel CHM. The Extent of Inflammatory Cell Infiltrate and Fibrosis in Lungs of Telomere- and Surfactant-Related Familial Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:736485. [PMID: 34631753 PMCID: PMC8497799 DOI: 10.3389/fmed.2021.736485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/27/2021] [Indexed: 12/04/2022] Open
Abstract
Familial pulmonary fibrosis (FPF) is a monogenic disease most commonly involving telomere- (TERT) or surfactant- (SFTP) related mutations. These mutations have been shown to alter lymphocytic inflammatory responses, and FPF biopsies with histological lymphocytic infiltrates have been reported. Recently, a model of a surfactant mutation in mice showed that the disease initially started with an inflammatory response followed by fibrogenesis. Since inflammation and fibrogenesis are targeted by different drugs, we investigated whether the degree of these two features co-localize or occur independently in different entities of FPF, and whether they influence survival. We quantified the number of lymphocyte aggregates per surface area, the extent of diffuse lymphocyte cell infiltrate, the number of fibroblast foci per surface area, and the percentage of fibrotic lung surface area in digitally scanned hematoxylin and eosin (H&E) sections of diagnostic surgical biopsies of patients with TERT-related FPF (TERT-PF; n = 17), SFTP-related FPF (SFTP-PF; n = 7), and sporadic idiopathic pulmonary fibrosis (sIPF; n = 10). For comparison, we included biopsies of patients with cellular non-specific interstitial pneumonia (cNSIP; n = 10), an inflammatory interstitial lung disease with high lymphocyte influx and usually responsive to immunosuppressive therapy. The degree of inflammatory cell infiltrate and fibrosis in TERT-PF and SFTP-PF was not significantly different from that in sIPF. In comparison with cNSIP, the extent of lymphocyte infiltrates was significantly lower in sIPF and TERT-PF, but not in SFTP-PF. However, in contrast with cNSIP, in sIPF, TERT-PF, and SFTP-PF, diffuse lymphocyte cell infiltrates were predominantly present and lymphocyte aggregates were only present in fibrotic areas (p < 0.0001). Furthermore, fibroblast foci and percentage of fibrotic lung surface were associated with survival (p = 0.022 and p = 0.018, respectively), while this association was not observed for lymphocyte aggregates or diffuse lymphocytic infiltration. Inflammatory cells in diagnostic lung biopsies of TERT-PF, SFTP-PF, and sIPF were largely confined to fibrotic areas. However, based on inflammation and fibrosis, no differences were found between FPF and sIPF, substantiating the histological similarities between monogenic familial and sporadic disease. Furthermore, the degree of fibrosis, rather than inflammation, correlates with survival, supporting that fibrogenesis is the key feature for therapeutic targeting of FPF.
Collapse
Affiliation(s)
- Aernoud A van Batenburg
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands
| | - Matthijs F M van Oosterhout
- Department of Pathology, DNA Pathology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands
| | - Sebastiaan N Knoppert
- Department of Pathology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Karin M Kazemier
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joanne J van der Vis
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands.,Department of Clinical Chemistry, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands
| | - Jan C Grutters
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Coline H M van Moorsel
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
26
|
Sjöblom N, Boyd S, Manninen A, Knuuttila A, Blom S, Färkkilä M, Arola J. Chronic cholestasis detection by a novel tool: automated analysis of cytokeratin 7-stained liver specimens. Diagn Pathol 2021; 16:41. [PMID: 33957930 PMCID: PMC8101247 DOI: 10.1186/s13000-021-01102-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/26/2021] [Indexed: 11/10/2022] Open
Abstract
Background The objective was to build a novel method for automated image analysis to locate and quantify the number of cytokeratin 7 (K7)-positive hepatocytes reflecting cholestasis by applying deep learning neural networks (AI model) in a cohort of 210 liver specimens. We aimed to study the correlation between the AI model’s results and disease progression. The cohort of liver biopsies which served as a model of chronic cholestatic liver disease comprised of patients diagnosed with primary sclerosing cholangitis (PSC). Methods In a cohort of patients with PSC identified from the PSC registry of the University Hospital of Helsinki, their K7-stained liver biopsy specimens were scored by a pathologist (human K7 score) and then digitally analyzed for K7-positive hepatocytes (K7%area). The digital analysis was by a K7-AI model created in an Aiforia Technologies cloud platform. For validation, values were human K7 score, stage of disease (Metavir and Nakunuma fibrosis score), and plasma liver enzymes indicating clinical cholestasis, all subjected to correlation analysis. Results The K7-AI model results (K7%area) correlated with the human K7 score (0.896; p < 2.2e− 16). In addition, K7%area correlated with stage of PSC (Metavir 0.446; p < 1.849e− 10 and Nakanuma 0.424; p < 4.23e− 10) and with plasma alkaline phosphatase (P-ALP) levels (0.369, p < 5.749e− 5). Conclusions The accuracy of the AI-based analysis was comparable to that of the human K7 score. Automated quantitative image analysis correlated with stage of PSC and with P-ALP. Based on the results of the K7-AI model, we recommend K7 staining in the assessment of cholestasis by means of automated methods that provide fast (9.75 s/specimen) quantitative analysis. Supplementary Information The online version contains supplementary material available at 10.1186/s13000-021-01102-6.
Collapse
Affiliation(s)
- Nelli Sjöblom
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, 00290, Helsinki, Finland.
| | - Sonja Boyd
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, 00290, Helsinki, Finland
| | - Anniina Manninen
- Aiforia Technologies Oy, Tukholmankatu 8, 000290, Helsinki, Finland
| | - Anna Knuuttila
- Aiforia Technologies Oy, Tukholmankatu 8, 000290, Helsinki, Finland
| | - Sami Blom
- Aiforia Technologies Oy, Tukholmankatu 8, 000290, Helsinki, Finland
| | - Martti Färkkilä
- Department of Gastroenterology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
| | - Johanna Arola
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, 00290, Helsinki, Finland
| |
Collapse
|
27
|
Malherbe K. Tumor Microenvironment and the Role of Artificial Intelligence in Breast Cancer Detection and Prognosis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1364-1373. [PMID: 33639101 DOI: 10.1016/j.ajpath.2021.01.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/02/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022]
Abstract
A critical knowledge gap has been noted in breast cancer detection, prognosis, and evaluation between tumor microenvironment and associated neoplasm. Artificial intelligence (AI) has multiple subsets or methods for data extraction and evaluation, including artificial neural networking, which allows computational foundations, similar to neurons, to make connections and new neural pathways during data set training. Deep machine learning and AI hold great potential to accurately assess tumor microenvironment models employing vast data management techniques. Despite the significant potential AI holds, there is still much debate surrounding the appropriate and ethical curation of medical data from picture archiving and communication systems. AI output's clinical significance depends on its human predecessor's data training sets. Integration between biomarkers, risk factors, and imaging data will allow the best predictor models for patient-based outcomes.
Collapse
Affiliation(s)
- Kathryn Malherbe
- Department Radiography, Faculty Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
28
|
Sivakumar P, Ammar R, Thompson JR, Luo Y, Streltsov D, Porteous M, McCoubrey C, Cantu E, Beers MF, Jarai G, Christie JD. Integrated plasma proteomics and lung transcriptomics reveal novel biomarkers in idiopathic pulmonary fibrosis. Respir Res 2021; 22:273. [PMID: 34689792 PMCID: PMC8543878 DOI: 10.1186/s12931-021-01860-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/09/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with a significant unmet medical need. Development of transformational therapies for IPF is challenging in part to due to lack of robust predictive biomarkers of prognosis and treatment response. Importantly, circulating biomarkers of IPF are limited and none are in clinical use. METHODS We previously reported dysregulated pathways and new disease biomarkers in advanced IPF through RNA sequencing of lung tissues from a cohort of transplant-stage IPF patients (n = 36) in comparison to normal healthy donors (n = 19) and patients with acute lung injury (n = 11). Here we performed proteomic profiling of matching plasma samples from these cohorts through the Somascan-1300 SomaLogics platform. RESULTS Comparative analyses of lung transcriptomic and plasma proteomic signatures identified a set of 34 differentially expressed analytes (fold change (FC) ≥ ± 1.5, false discovery ratio (FDR) ≤ 0.1) in IPF samples compared to healthy controls. IPF samples showed strong enrichment of chemotaxis, tumor infiltration and mast cell migration pathways and downregulated extracellular matrix (ECM) degradation. Mucosal (CCL25 and CCL28) and Th2 (CCL17 and CCL22) chemokines were markedly upregulated in IPF and highly correlated within the subjects. The mast cell maturation chemokine, CXCL12, was also upregulated in IPF plasma (fold change 1.92, FDR 0.006) and significantly correlated (Pearson r = - 0.38, p = 0.022) to lung function (%predicted FVC), with a concomitant increase in the mast cell Tryptase, TPSB2. Markers of collagen III and VI degradation (C3M and C6M) were significantly downregulated (C3M p < 0.001 and C6M p < 0.0001 IPF vs control) and correlated, Pearson r = 0.77) in advanced IPF consistent with altered ECM homeostasis. CONCLUSIONS Our study identifies a panel of tissue and circulating biomarkers with clinical utility in IPF that can be validated in future studies across larger cohorts.
Collapse
Affiliation(s)
- Pitchumani Sivakumar
- grid.419971.30000 0004 0374 8313Translational Early Development, Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Road, Princeton, NJ 08540 USA
| | - Ron Ammar
- grid.419971.30000 0004 0374 8313Informatics and Predictive Sciences, Bristol-Myers Squibb Research and Development, Princeton, NJ USA
| | - John Ryan Thompson
- grid.419971.30000 0004 0374 8313Informatics and Predictive Sciences, Bristol-Myers Squibb Research and Development, Princeton, NJ USA
| | - Yi Luo
- grid.419971.30000 0004 0374 8313Translational Medicine, Bristol-Myers Squibb Research and Development, Princeton, NJ USA
| | - Denis Streltsov
- grid.419971.30000 0004 0374 8313Fibrosis Discovery Biology, Bristol-Myers Squibb Research and Development, Princeton, NJ USA
| | - Mary Porteous
- grid.25879.310000 0004 1936 8972Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Carly McCoubrey
- grid.25879.310000 0004 1936 8972Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Edward Cantu
- grid.25879.310000 0004 1936 8972Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania, Philadelphia, PA USA
| | - Michael F. Beers
- grid.25879.310000 0004 1936 8972Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972PENN Lung Biology Institute, University of Pennsylvania, Philadelphia, PA USA
| | - Gabor Jarai
- grid.419971.30000 0004 0374 8313Fibrosis Discovery Biology, Bristol-Myers Squibb Research and Development, Princeton, NJ USA
| | - Jason D. Christie
- grid.25879.310000 0004 1936 8972Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972PENN Lung Biology Institute, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|