1
|
Tocci P, Roman C, Sestito R, Caprara V, Sacconi A, Molineris I, Tonon G, Blandino G, Bagnato A. The endothelin-1-driven tumor-stroma feed-forward loops in high-grade serous ovarian cancer. Clin Sci (Lond) 2024; 138:851-862. [PMID: 38884602 PMCID: PMC11230866 DOI: 10.1042/cs20240346] [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: 02/23/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/18/2024]
Abstract
The high-grade serous ovarian cancer (HG-SOC) tumor microenvironment (TME) is constellated by cellular elements and a network of soluble constituents that contribute to tumor progression. In the multitude of the secreted molecules, the endothelin-1 (ET-1) has emerged to be implicated in the tumor/TME interplay; however, the molecular mechanisms induced by the ET-1-driven feed-forward loops (FFL) and associated with the HG-SOC metastatic potential need to be further investigated. The tracking of the patient-derived (PD) HG-SOC cell transcriptome by RNA-seq identified the vascular endothelial growth factor (VEGF) gene and its associated signature among those mostly up-regulated by ET-1 and down-modulated by the dual ET-1R antagonist macitentan. Within the ligand-receptor pairs concurrently expressed in PD-HG-SOC cells, endothelial cells and activated fibroblasts, we discovered two intertwined FFL, the ET-1/ET-1R and VEGF/VEGF receptors, concurrently activated by ET-1 and shutting-down by macitentan, or by the anti-VEGF antibody bevacizumab. In parallel, we observed that ET-1 fine-tuned the tumoral and stromal secretome toward a pro-invasive pattern. Into the fray of the HG-SOC/TME double and triple co-cultures, the secretion of ET-1 and VEGF, that share a common co-regulation, was inhibited upon the administration of macitentan. Functionally, macitentan, mimicking the effect of bevacizumab, interfered with the HG-SOC/TME FFL-driven communication that fuels the HG-SOC invasive behavior. The identification of ET-1 and VEGF FFL as tumor and TME actionable vulnerabilities, reveals how ET-1R blockade, targeting the HG-SOC cells and the TME simultaneously, may represent an effective therapeutic option for HG-SOC patients.
Collapse
Affiliation(s)
- Piera Tocci
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Celia Roman
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Rosanna Sestito
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Valentina Caprara
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Sacconi
- Translational Oncology Research Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Ivan Molineris
- Department of Life Science and System Biology, University of Turin, Turin, Italy
| | - Giovanni Tonon
- Center for Omics Sciences (COSR) and Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
2
|
Xu AM, Haro M, Walts AE, Hu Y, John J, Karlan BY, Merchant A, Orsulic S. Spatiotemporal architecture of immune cells and cancer-associated fibroblasts in high-grade serous ovarian carcinoma. SCIENCE ADVANCES 2024; 10:eadk8805. [PMID: 38630822 PMCID: PMC11023532 DOI: 10.1126/sciadv.adk8805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
High-grade serous ovarian carcinoma (HGSOC), the deadliest form of ovarian cancer, is typically diagnosed after it has metastasized and often relapses after standard-of-care platinum-based chemotherapy, likely due to advanced tumor stage, heterogeneity, and immune evasion and tumor-promoting signaling from the tumor microenvironment. To understand how spatial heterogeneity contributes to HGSOC progression and early relapse, we profiled an HGSOC tissue microarray of patient-matched longitudinal samples from 42 patients. We found spatial patterns associated with early relapse, including changes in T cell localization, malformed tertiary lymphoid structure (TLS)-like aggregates, and increased podoplanin-positive cancer-associated fibroblasts (CAFs). Using spatial features to compartmentalize the tissue, we found that plasma cells distribute in two different compartments associated with TLS-like aggregates and CAFs, and these distinct microenvironments may account for the conflicting reports about the role of plasma cells in HGSOC prognosis.
Collapse
Affiliation(s)
- Alexander M. Xu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marcela Haro
- Department of Obstetrics and Gynecology and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ye Hu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Joshi John
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Department of Medicine, Division of Geriatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Akil Merchant
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
van Wagensveld L, Walker C, Hahn K, Sanders J, Kruitwagen R, van der Aa M, Sonke G, Rottenberg S, de Vijver KV, Janowczyk A, Horlings H. The prognostic value of tumor-stroma ratio and a newly developed computer-aided quantitative analysis of routine H&E slides in high-grade serous ovarian cancer. RESEARCH SQUARE 2023:rs.3.rs-3511087. [PMID: 38014112 PMCID: PMC10680933 DOI: 10.21203/rs.3.rs-3511087/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
INTRODUCTION Tumor-stroma ratio (TSR) is prognostic in multiple cancers, while its role in high-grade serous ovarian cancer (HGSOC) remains unclear. Despite the prognostic insight gained from genetic profiles and tumor-infiltrating lymphocytes (TILs), the prognostic use of histology slides remains limited, while it enables the identification of tumor characteristics via computational pathology reducing scoring time and costs. To address this, this study aimed to assess TSR's prognostic role in HGSOC and its association with TILs. We additionally developed an algorithm, Ovarian-TSR (OTSR), using deep learning for TSR scoring, comparing it to manual scoring. Methods : 340 patients with advanced-stage who underwent primary debulking surgery (PDS) or neo-adjuvant chemotherapy (NACT) with interval debulking (IDS). TSR was assessed in both the most invasive (MI) and whole tumor (WT) regions through manual scoring by pathologists and quantification using OTSR. Patients were categorized as stroma-rich (≥ 50% stroma) or stroma-poor (< 50%). TILs were evaluated via immunohistochemical staining. Result s: In PDS, stroma-rich tumors were significantly associated with a more frequent papillary growth pattern (60% vs 34%), while In NACT stroma-rich tumors had a lower Tumor Regression Grading (TRG 4&5, 21% vs 57%) and increased pleural metastasis (25% vs 16%). Stroma-rich patients had significantly shorter overall and progression-free survival compared to stroma-poor (31 versus 45 months; P < 0.0001, and 15 versus 17 months; P = 0.0008, respectively). Combining stromal percentage and TILs led to three distinct survival groups with good (stroma-poor, high TIL), medium (stroma-rich, high TIL, or; stroma-poor, Low TIL), and poor(stroma-rich, low TIL) survival. These survival groups remained significant in CD8 and CD103 in multivariable analysis (Hazard ratio (HR) = 1.42, 95% Confidence-interval (CI) = 1.02-1.99; HR = 1.49, 95% CI = 1.01-2.18, and HR = 1.48, 95% CI = 1.05-2.08; HR = 2.24, 95% CI = 1.55-3.23, respectively). OTSR was able to recapitulate these results and demonstrated high concordance with expert pathologists (correlation = 0.83). Conclusions : TSR is an independent prognostic factor for survival assessment in HGSOC. Stroma-rich tumors have a worse prognosis and, in the case of NACT, a higher likelihood of pleural metastasis. OTSR provides a cost and time-efficient way of determining TSR with high reproducibility and reduced inter-observer variability.
Collapse
|
4
|
Li YR, Ochoa CJ, Zhu Y, Kramer A, Wilson M, Fang Y, Chen Y, Singh T, Di Bernardo G, Zhu E, Lee D, Moatamed NA, Bando J, Zhou JJ, Memarzadeh S, Yang L. Profiling ovarian cancer tumor and microenvironment during disease progression for cell-based immunotherapy design. iScience 2023; 26:107952. [PMID: 37810241 PMCID: PMC10558812 DOI: 10.1016/j.isci.2023.107952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
Ovarian cancer (OC) is highly lethal due to late detection and frequent recurrence. Initial treatments, comprising surgery and chemotherapy, lead to disease remission but are invariably associated with subsequent relapse. The identification of novel therapies and an improved understanding of the molecular and cellular characteristics of OC are urgently needed. Here, we conducted a comprehensive analysis of primary tumor cells and their microenvironment from 16 chemonaive and 10 recurrent OC patient samples. Profiling OC tumor biomarkers allowed for the identification of potential molecular targets for developing immunotherapies, while profiling the microenvironment yielded insights into its cellular composition and property changes between chemonaive and recurrent samples. Notably, we identified CD1d as a biomarker of the OC microenvironment and demonstrated its targeting by invariant natural killer T (iNKT) cells. Overall, our study presents a comprehensive immuno-profiling of OC tumor and microenvironment during disease progression, guiding the development of immunotherapies for OC treatment, especially for recurrent disease.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher J Ochoa
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam Kramer
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew Wilson
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tanya Singh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gabriella Di Bernardo
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Enbo Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Derek Lee
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Neda A Moatamed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joanne Bando
- Department of Medicine, Division of Pulmonary and Critical Care, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin J Zhou
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Ferri-Borgogno S, Zhu Y, Sheng J, Burks JK, Gomez JA, Wong KK, Wong ST, Mok SC. Spatial Transcriptomics Depict Ligand-Receptor Cross-talk Heterogeneity at the Tumor-Stroma Interface in Long-Term Ovarian Cancer Survivors. Cancer Res 2023; 83:1503-1516. [PMID: 36787106 PMCID: PMC10159916 DOI: 10.1158/0008-5472.can-22-1821] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/06/2022] [Accepted: 02/10/2023] [Indexed: 02/15/2023]
Abstract
Advanced high-grade serous ovarian cancer (HGSC) is an aggressive disease that accounts for 70% of all ovarian cancer deaths. Nevertheless, 15% of patients diagnosed with advanced HGSC survive more than 10 years. The elucidation of predictive markers of these long-term survivors (LTS) could help identify therapeutic targets for the disease, and thus improve patient survival rates. To investigate the stromal heterogeneity of the tumor microenvironment (TME) in ovarian cancer, we used spatial transcriptomics to generate spatially resolved transcript profiles in treatment-naïve advanced HGSC from LTS and short-term survivors (STS) and determined the association between cancer-associated fibroblasts (CAF) heterogeneity and survival in patients with advanced HGSC. Spatial transcriptomics and single-cell RNA-sequencing data were integrated to distinguish tumor and stroma regions, and a computational method was developed to investigate spatially resolved ligand-receptor interactions between various tumor and CAF subtypes in the TME. A specific subtype of CAFs and its spatial location relative to a particular ovarian cancer cell subtype in the TME correlated with long-term survival in patients with advanced HGSC. Also, increased APOE-LRP5 cross-talk occurred at the stroma-tumor interface in tumor tissues from STS compared with LTS. These findings were validated using multiplex IHC. Overall, this spatial transcriptomics analysis revealed spatially resolved CAF-tumor cross-talk signaling networks in the ovarian TME that are associated with long-term survival of patients with HGSC. Further studies to confirm whether such cross-talk plays a role in modulating the malignant phenotype of HGSC and could serve as a predictive biomarker of patient survival are warranted. SIGNIFICANCE Generation of spatially resolved gene expression patterns in tumors from patients with ovarian cancer surviving more than 10 years allows the identification of novel predictive biomarkers and therapeutic targets for better patient management. See related commentary by Kelliher and Lengyel, p. 1383.
Collapse
Affiliation(s)
- Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying Zhu
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Jianting Sheng
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Javier A. Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kwong Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen T.C. Wong
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Samuel C. Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
6
|
Orsulic S, John J, Walts AE, Gertych A. Computational pathology in ovarian cancer. Front Oncol 2022; 12:924945. [PMID: 35965569 PMCID: PMC9372445 DOI: 10.3389/fonc.2022.924945] [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: 04/20/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Histopathologic evaluations of tissue sections are key to diagnosing and managing ovarian cancer. Pathologists empirically assess and integrate visual information, such as cellular density, nuclear atypia, mitotic figures, architectural growth patterns, and higher-order patterns, to determine the tumor type and grade, which guides oncologists in selecting appropriate treatment options. Latent data embedded in pathology slides can be extracted using computational imaging. Computers can analyze digital slide images to simultaneously quantify thousands of features, some of which are visible with a manual microscope, such as nuclear size and shape, while others, such as entropy, eccentricity, and fractal dimensions, are quantitatively beyond the grasp of the human mind. Applications of artificial intelligence and machine learning tools to interpret digital image data provide new opportunities to explore and quantify the spatial organization of tissues, cells, and subcellular structures. In comparison to genomic, epigenomic, transcriptomic, and proteomic patterns, morphologic and spatial patterns are expected to be more informative as quantitative biomarkers of complex and dynamic tumor biology. As computational pathology is not limited to visual data, nuanced subvisual alterations that occur in the seemingly “normal” pre-cancer microenvironment could facilitate research in early cancer detection and prevention. Currently, efforts to maximize the utility of computational pathology are focused on integrating image data with other -omics platforms that lack spatial information, thereby providing a new way to relate the molecular, spatial, and microenvironmental characteristics of cancer. Despite a dire need for improvements in ovarian cancer prevention, early detection, and treatment, the ovarian cancer field has lagged behind other cancers in the application of computational pathology. The intent of this review is to encourage ovarian cancer research teams to apply existing and/or develop additional tools in computational pathology for ovarian cancer and actively contribute to advancing this important field.
Collapse
Affiliation(s)
- Sandra Orsulic
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, United States
- *Correspondence: Sandra Orsulic,
| | - Joshi John
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Arkadiusz Gertych
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Faculty of Biomedical Engineering, Silesian University of Technology, Zabrze, Poland
| |
Collapse
|
7
|
Osteosarcoma Detection from Whole Slide Images Using Multi-Feature Non-Seed-Based Region Growing Segmentation and Feature Extraction. Neural Process Lett 2022. [DOI: 10.1007/s11063-022-10914-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
8
|
Horst EN, Novak CM, Burkhard K, Snyder CS, Verma R, Crochran DE, Geza IA, Fermanich W, Mehta P, Schlautman DC, Tran LA, Brezenger ME, Mehta G. Injectable three-dimensional tumor microenvironments to study mechanobiology in ovarian cancer. Acta Biomater 2022; 146:222-234. [PMID: 35487424 PMCID: PMC10538942 DOI: 10.1016/j.actbio.2022.04.039] [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: 08/30/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022]
Abstract
Epithelial ovarian cancers are among the most aggressive forms of gynecological malignancies. Despite the advent of poly adenosine diphosphate-ribose polymerase (PARP) and checkpoint inhibitors, improvement to patient survival has been modest. Limited in part by clinical translation, beneficial therapeutic strategies remain elusive in ovarian cancers. Although elevated levels of extracellular proteins, including collagens, proteoglycans, and glycoproteins, have been linked to chemoresistance, they are often missing from the processes of drug- development and screening. Biophysical and biochemical signaling from the extracellular matrix (ECM) determine cellular phenotype and affect both tumor progression and therapeutic response. However, many state-of-the-art tumor models fail to mimic the complexities of the tumor microenvironment (TME) and omit key signaling components. In this article, two interpenetrating network (IPN) hydrogel scaffold platforms, comprising of alginate-collagen or agarose-collagen, have been characterized for use as 3D in vitro models of epithelial ovarian cancer ECM. These highly tunable, injection mold compatible, and inexpensive IPNs replicate the critical governing physical and chemical signaling present within the ovarian TME. Additionally, an effective and cell-friendly live-cell retrieval method has been established to recover cells post-encapsulation. Lastly, functional mechanotransduction in ovarian cancers was demonstrated by increasing scaffold stiffness within the 3D in vitro ECM models. With these features, the agarose-collagen and alginate-collagen hydrogels provide a robust TME for the study of mechanobiology in epithelial cancers. STATEMENT OF SIGNIFICANCE: Ovarian cancer is the most lethal gynecologic cancer afflicting women today. Here we present the development, characterization, and validation of 3D interpenetrating platforms to shift the paradigm in standard in vitro modeling. These models help elucidate the roles of biophysical and biochemical cues in ovarian cancer progression. The agarose-collagen and alginate-collagen interpenetrating network (IPN) hydrogels are simple to fabricate, inexpensive, and can be modified to create custom mechanical stiffnesses and concentrations of bio-adhesive motifs. Given that investigations into the roles of biophysical characteristics in ovarian cancers have provided incongruent results, we believe that the IPN platforms will be critically important to uncovering molecular drivers. We also expect these platforms to be broadly applicable to studies involving mechanobiology in solid tumors.
Collapse
Affiliation(s)
- Eric N Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Caymen M Novak
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States
| | - Kathleen Burkhard
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Catherine S Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Rhea Verma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Darel E Crochran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Izabella A Geza
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Wesley Fermanich
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Denise C Schlautman
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Linh A Tran
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Michael E Brezenger
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, United States; Precision Health, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
9
|
M A M, Kim JY, Pan CH, Kim E. The impact of the spatial heterogeneity of resistant cells and fibroblasts on treatment response. PLoS Comput Biol 2022; 18:e1009919. [PMID: 35263336 PMCID: PMC8906648 DOI: 10.1371/journal.pcbi.1009919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
A long-standing practice in the treatment of cancer is that of hitting hard with the maximum tolerated dose to eradicate tumors. This continuous therapy, however, selects for resistant cells, leading to the failure of the treatment. A different type of treatment strategy, adaptive therapy, has recently been shown to have a degree of success in both preclinical xenograft experiments and clinical trials. Adaptive therapy is used to maintain a tumor's volume by exploiting the competition between drug-sensitive and drug-resistant cells with minimum effective drug doses or timed drug holidays. To further understand the role of competition in the outcomes of adaptive therapy, we developed a 2D on-lattice agent-based model. Our simulations show that the superiority of the adaptive strategy over continuous therapy depends on the local competition shaped by the spatial distribution of resistant cells. Intratumor competition can also be affected by fibroblasts, which produce microenvironmental factors that promote cancer cell growth. To this end, we simulated the impact of different fibroblast distributions on treatment outcomes. As a proof of principle, we focused on five types of distribution of fibroblasts characterized by different locations, shapes, and orientations of the fibroblast region with respect to the resistant cells. Our simulation shows that the spatial architecture of fibroblasts modulates tumor progression in both continuous and adaptive therapy. Finally, as a proof of concept, we simulated the outcomes of adaptive therapy of a virtual patient with four metastatic sites composed of different spatial distributions of fibroblasts and drug-resistant cell populations. Our simulation highlights the importance of undetected metastatic lesions on adaptive therapy outcomes.
Collapse
Affiliation(s)
- Masud M A
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| |
Collapse
|
10
|
Corvo A, Caballero HSG, Westenberg MA, van Driel MA, van Wijk JJ. Visual Analytics for Hypothesis-Driven Exploration in Computational Pathology. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2021; 27:3851-3866. [PMID: 32340951 DOI: 10.1109/tvcg.2020.2990336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Recent advances in computational and algorithmic power are evolving the field of medical imaging rapidly. In cancer research, many new directions are sought to characterize patients with additional imaging features derived from radiology and pathology images. The emerging field of Computational Pathology targets the high-throughput extraction and analysis of the spatial distribution of cells from digital histopathology images. The associated morphological and architectural features allow researchers to quantify and characterize new imaging biomarkers for cancer diagnosis, prognosis, and treatment decisions. However, while the image feature space grows, exploration and analysis become more difficult and ineffective. There is a need for dedicated interfaces for interactive data manipulation and visual analysis of computational pathology and clinical data. For this purpose, we present IIComPath, a visual analytics approach that enables clinical researchers to formulate hypotheses and create computational pathology pipelines involving cohort construction, spatial analysis of image-derived features, and cohort analysis. We demonstrate our approach through use cases that investigate the prognostic value of current diagnostic features and new computational pathology biomarkers.
Collapse
|
11
|
Jiang J, Tekin B, Guo R, Liu H, Huang Y, Wang C. Digital Pathology-based Study of Cell- and Tissue-level Morphologic Features in Serous Borderline Ovarian Tumor and High-grade Serous Ovarian Cancer. J Pathol Inform 2021; 12:24. [PMID: 34447604 PMCID: PMC8356706 DOI: 10.4103/jpi.jpi_76_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/28/2020] [Accepted: 02/11/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Serous borderline ovarian tumor (SBOT) and high-grade serous ovarian cancer (HGSOC) are two distinct subtypes of epithelial ovarian tumors, with markedly different biologic background, behavior, prognosis, and treatment. However, the histologic diagnosis of serous ovarian tumors can be subjectively variable and labor-intensive as multiple tumor slides/blocks need to be thoroughly examined to search for these features. Materials and Methods: We developed a novel informatics system to facilitate objective and scalable diagnosis screening for SBOT and HGSOC. The system was built upon Groovy scripts and QuPath to enable interactive annotation and data exchange. Results: The system was used to successfully detect cellular boundaries and extract an expanded set of cellular features representing cell- and tissue-level characteristics. The performance of cell-level classification for both tumor and stroma cells achieved >90% accuracy. The performance of differentiating HGSOC versus SBOT achieved 91%–95% accuracy for 6485 imaging patches which have sufficient tumor and stroma cells (minimum of ten each) and 97% accuracy for classifying patients when aggregating the results to whole-slide image based on consensus. Conclusions: Cellular features digitally extracted from pathological images can be used for cell classification and SBOT v. HGSOC differentiation. Introducing digital pathology into ovarian cancer research could be beneficial to discover potential clinical implications. A larger cohort is required to further evaluate the system.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Burak Tekin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ruifeng Guo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Yajue Huang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Chen Wang
- Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
12
|
Wei W, Zheng L, Gao Y, He M, Yang F. Expression and prognostic significance of NKD2 in ovarian cancer. Jpn J Clin Oncol 2021; 51:459-468. [PMID: 33324989 DOI: 10.1093/jjco/hyaa244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/23/2020] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Naked2 (NKD2) is a negative regulator of Wnt signaling pathway and associates with transforming growth factor secretion. The role of NKD2 in ovarian cancer is unknown. PATIENTS AND METHODS Gene expression profiles were measured and compared in nine patients by RNA sequencing. NKD2 expressions in ovarian cancer were measured by reverse transcription polymerase chain reaction and western blot. Tissue slides of 79 patients were stained and scored for NKD2 expression. In vitro experiments were conducted to explore the role of NKD2 in ovarian cancer. The prognostic role of NKD2 was evaluated by survival analysis. RESULTS NKD2 was upregulated in patients with better survival by mRNA and protein expression. Patients were classified as NKD2-high group (n = 30) and NKD2-low group (n = 49) according to immunohistochemical score. High NKD2 was correlated with lower recurrence rate (P = 0.002) and higher percentage of platinum-sensitive recurrence (P = 0.006). Median progression-free survival was significantly longer for NKD2-high patients than NKD2-low patients (49.1 vs.14.1 months, P < 0.001). Accordingly, there was a significantly difference in terms of overall survival time between two groups (hazard ratio: 3.04; 95% confidence interval: 1.58-5.85, P < 0.001). Multivariate regression suggested that NKD2 was independently prognostic factors in terms of progression-free survival (hazard ratio: 2.91; 95% confidence interval: 1.61-5.27, P < 0.001) and overall survival (hazard ratio: 3.6; 95% confidence interval: 1.80-7.21, P < 0.001). In vitro studies further demonstrated that NKD2 suppressed ovarian cancer cell proliferation, colony formation and cell migration. CONCLUSION NKD2 is a novel prognostic marker and could suppress tumor progression in ovarian cancer.
Collapse
Affiliation(s)
- Wei Wei
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, China, China
| | - Lisi Zheng
- Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen, University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, China
| | - Ying Gao
- Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen, University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, China
| | - Minjun He
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, China, China
| | - Fan Yang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, China, China
| |
Collapse
|
13
|
Li M, Bai J, Wang S, Zhai Y, Zhang S, Li C, Du J, Zhang Y. Clinical Implication of Systemic Immune-Inflammation Index and Prognostic Nutritional Index in Skull Base Chordoma Patients. Front Oncol 2021; 11:548325. [PMID: 33718126 PMCID: PMC7947628 DOI: 10.3389/fonc.2021.548325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 02/04/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammation associated markers and nutritional indexes are associated with survival, and act as novel prognostic grading systems in patients with cancer, though the role of these markers in chordoma remains unclear. The current study aimed to characterize systemic immune-inflammation index (SII) and prognostic nutritional index (PNI), and their relationship with clinicopathological data and survival in skull base chordoma. Our retrospective study enrolled 183 patients with primary skull base chordoma who received surgical treatment. Clinicopathological data and preoperative blood tests including neutrophil, lymphocyte, platelet counts and albumin level were collected from medical records. Neutrophil lymphocyte ratio (NLR), platelet lymphocyte ratio (PLR), SII, PNI were calculated and the optimal cut-off values of these markers were used for further survival analysis via Kaplan–Meier survival analysis and Cox proportional hazards regression analysis. The value of NLR, PLR, SII, and PNI in skull base chordoma ranged from 0.44–6.48, 45.36–273.94, 113.37–1761.45, and 43.40–70.65, respectively. PNI was significantly correlated with patients' sex (p = 0.005) and age (p = 0.037). SII was positively correlated with NLR and PLR, but negatively correlated with PNI. The median overall survival (OS) time was 74.0 months and Kaplan–Meier survival analysis indicated that all four indexes were associated with OS. Multivariable Cox proportional hazards regression analysis identified that high SII was an independent prognostic factor for poor OS. More importantly, patients with high SII and PNI had the worst outcomes and combined use of SII and PNI increased the predictive ability for patients' survival in skull base chordoma. Our results suggest SII and PNI may be effective prognostic indicators of OS for patients with primary skull base chordoma after surgical resection.
Collapse
Affiliation(s)
- Mingxuan Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yixuan Zhai
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuheng Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiang Du
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Key Laboratory of Central Nervous System Injury Research, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Zou MX, Zheng BW, Liu FS, Wang XB, Hu JR, Huang W, Dai ZH, Zhang QS, Liu FB, Zhong H, Jiang Y, She XL, Li XB, Lv GH, Li J. The Relationship Between Tumor-Stroma Ratio, the Immune Microenvironment, and Survival in Patients With Spinal Chordoma. Neurosurgery 2020; 85:E1095-E1110. [PMID: 31501892 DOI: 10.1093/neuros/nyz333] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Currently, little is known about the clinical relevance of tumor-stroma ratio (TSR) in chordoma and data discussing the relationship between TSR and immune status of chordoma are lacking. OBJECTIVE To characterize TSR distribution in spinal chordoma, and investigated its correlation with clinicopathologic or immunological features of patients and outcome. METHODS TSR was assessed visually on hematoxylin and eosin-stained sections from 54 tumor specimens by 2 independent pathologists. Multiplex immunofluorescence was used to quantify the expression levels of microvessel density, Ki-67, Brachyury, and tumor as well as stromal PD-L1. Tumor immunity status including the Immunoscore and densities of tumor-infiltrating lymphocytes (TILs) subtypes were obtained from our published data and reanalyzed. RESULTS Bland-Altman plot showed no difference between mean TSR derived from the two observers. TSR was positively associated with stromal PD-L1 expression, the Immunoscore and CD3+ as well as CD4+ TILs density, but negatively correlated with tumor microvessel density, Ki-67 index, surrounding muscle invasion by tumor and number of Foxp3+ and PD-1+ TILs. Low TSR independently predicted poor local recurrence-free survival and overall survival. Moreover, patients with low TSR and low Immunoscore chordoma phenotype were associated with the worst survival. More importantly, combined TSR and Immunoscore accurately reflected prognosis and enhanced the ability of TSR or Immunoscore alone for outcome prediction. CONCLUSION These data reveal the significant impact of TSR on tumor progression and immunological response of patients. Subsequent use of agents targeting the stroma compartment may be an effective strategy to treat chordoma especially in combination with immune-based drugs.
Collapse
Affiliation(s)
- Ming-Xiang Zou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Bo-Wen Zheng
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Fu-Sheng Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Jia-Rui Hu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Wei Huang
- Institute of Precision Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhe-Hao Dai
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Qian-Shi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Fu-Bing Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Hua Zhong
- Department of Orthopedics Surgery, Central Hospital of Yi Yang, Yiyang, China
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Xiao-Ling She
- Department of Pathology, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Xiao-Bing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Guo-Hua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| |
Collapse
|
15
|
Jiang X, Tomlinson IPM. Why is cancer not more common? A changing microenvironment may help to explain why, and suggests strategies for anti-cancer therapy. Open Biol 2020; 10:190297. [PMID: 32289242 PMCID: PMC7241076 DOI: 10.1098/rsob.190297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/25/2020] [Indexed: 12/27/2022] Open
Abstract
One of the great unsolved puzzles in cancer biology is not why cancers occur, but rather explaining why so few cancers occur compared with the theoretical number that could occur, given the number of progenitor cells in the body and the normal mutation rate. We hypothesized that a contributory explanation is that the tumour microenvironment (TME) is not fixed due to factors such as immune cell infiltration, and that this could impair the ability of neoplastic cells to retain a high enough fitness to become a cancer. The TME has implicitly been assumed to be static in most cancer evolution models, and we therefore developed a mathematical model of spatial cancer evolution assuming that the TME, and thus the optimum cancer phenotype, changes over time. Based on simulations, we show how cancer cell populations adapt to diverse changing TME conditions and fitness landscapes. Compared with static TMEs, which generate neutral dynamics, changing TMEs lead to complex adaptations with characteristic spatio-temporal heterogeneity involving variable fitness effects of driver mutations, subclonal mixing, subclonal competition and phylogeny patterns. In many cases, cancer cell populations fail to grow or undergo spontaneous regression, and even extinction. Our analyses predict that cancer evolution in a changing TME is challenging, and can help to explain why cancer is neither inevitable nor as common as expected. Should cancer driver mutations with effects dependent of the TME exist, they are likely to be selected. Anti-cancer prevention and treatment strategies based on changing the TME are feasible and potentially effective.
Collapse
Affiliation(s)
| | - Ian P. M. Tomlinson
- Edinburgh Cancer Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| |
Collapse
|
16
|
Singh S, Ray LA, Shahi Thakuri P, Tran S, Konopka MC, Luker GD, Tavana H. Organotypic breast tumor model elucidates dynamic remodeling of tumor microenvironment. Biomaterials 2020; 238:119853. [PMID: 32062146 PMCID: PMC8165649 DOI: 10.1016/j.biomaterials.2020.119853] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/13/2022]
Abstract
Fibroblasts are a critical component of tumor microenvironments and associate with cancer cells physically and biochemically during different stages of the disease. Existing cell culture models to study interactions between fibroblasts and cancer cells lack native tumor architecture or scalability. We developed a scalable organotypic model by robotically encapsulating a triple negative breast cancer (TNBC) cell spheroid within a natural extracellular matrix containing dispersed fibroblasts. We utilized an established CXCL12 - CXCR4 chemokine-receptor signaling in breast tumors to validate our model. Using imaging techniques and molecular analyses, we demonstrated that CXCL12-secreting fibroblasts have elevated activity of RhoA/ROCK/myosin light chain-2 pathway and rapidly and significantly contract collagen matrices. Signaling between TNBC cells and CXCL12-producing fibroblasts promoted matrix invasion of cancer cells by activating oncogenic mitogen-activated protein kinase signaling, whereas normal fibroblasts significantly diminished TNBC cell invasiveness. We demonstrated that disrupting CXCL12 - CXCR4 signaling using a molecular inhibitor significantly inhibited invasiveness of cancer cells, suggesting blocking of tumor-stromal interactions as a therapeutic strategy especially for cancers such as TNBC that lack targeted therapies. Our organotypic tumor model mimics native solid tumors, enables modular addition of different stromal cells and extracellular matrix proteins, and allows high throughput compound screening against tumor-stromal interactions to identify novel therapeutics.
Collapse
Affiliation(s)
- Sunil Singh
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Lucille A Ray
- Department of Chemistry, The University of Akron, Akron, OH, 44325, USA
| | - Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Sydnie Tran
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Michael C Konopka
- Department of Chemistry, The University of Akron, Akron, OH, 44325, USA
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA.
| |
Collapse
|
17
|
Zou M, Pan Y, Huang W, Zhang T, Escobar D, Wang X, Jiang Y, She X, Lv G, Li J. A four-factor immune risk score signature predicts the clinical outcome of patients with spinal chordoma. Clin Transl Med 2020; 10:224-237. [PMID: 32508056 PMCID: PMC7240847 DOI: 10.1002/ctm2.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Currently, the measurement of immune cells in previous studies is usually subjective, and no immune-based prognostic model has been established for chordoma. In this study, we sought to simultaneously measure tumor-infiltrating lymphocyte (TIL) subtypes in chordoma samples using an objective method and develop an immune risk score (IRS) model for survival prediction. METHODS Multiplexed quantitative immunofluorescence staining was used to determine the TIL levels in the tumoral and stromal subareas of 114 spinal chordoma specimens (54 in the training and 60 in the validation cohort) for programmed death-1 (PD-1), CD3, CD8, CD20 (where CD is cluster of differentiation), and FOXP3. Flow cytometry was performed to validate the immunofluorescence assay for lymphocyte measurement on an additional five fresh chordoma specimens. Subsequently, the IRS model was built using the least absolute shrinkage and selection operator (LASSO) Cox regression method. RESULTS Flow cytometry and quantitative immunofluorescence showed similar lymphocytic percentages and TIL subpopulation proportions in the fresh tumor specimens. With the training data, the LASSO model identified four immune features for IRS construction: tumoral FOXP3, tumoral PD-1, stromal FOXP3, and stromal CD8. In both cohorts, a high IRS was significantly associated with tumoral programmed cell death-1 ligand 1 expression, Enneking inappropriate tumor resection, and surrounding muscle invasion by tumor. Multivariate Cox regression and stratified analysis in the two cohorts revealed that the IRS was an independent predictor and could effectively separate patients with similar Enneking staging into different risk subgroups, with significantly different survival rates. Further receiver operating characteristic analysis found that the IRS classifier had a better prognostic value than the traditional clinicopathological factors and compensated for the deficiency of Enneking staging for outcome prediction. More importantly, a nomogram based on the IRS and clinical predictors showed adequate performance in estimating disease recurrence and survival of patients. CONCLUSIONS These data support the use of the IRS signature as a reliable prognostic tool in spinal chordoma and may facilitate individualized therapy decision making for patients.
Collapse
Affiliation(s)
- Ming‐Xiang Zou
- Department of Spine SurgeryThe First Affiliated HospitalUniversity of South ChinaHengyangChina
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Yue Pan
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Wei Huang
- Institute of Precision MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Tao‐Lan Zhang
- Department of Cancer BiologyCollege of Medicine & Life SciencesUniversity of ToledoToledoOhio
| | - David Escobar
- Department of Cancer BiologyCollege of Medicine & Life SciencesUniversity of ToledoToledoOhio
| | - Xiao‐Bin Wang
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Yi Jiang
- Department of PathologyThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiao‐Ling She
- Department of PathologyThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Guo‐Hua Lv
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Jing Li
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
18
|
Liu K, Xia W, Qiang M, Chen X, Liu J, Guo X, Lv X. Deep learning pathological microscopic features in endemic nasopharyngeal cancer: Prognostic value and protentional role for individual induction chemotherapy. Cancer Med 2019; 9:1298-1306. [PMID: 31860791 PMCID: PMC7013063 DOI: 10.1002/cam4.2802] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND To explore the prognostic value and the role for treatment decision of pathological microscopic features in patients with nasopharyngeal carcinoma (NPC) using the method of deep learning. METHODS The pathological microscopic features were extracted using the software QuPath (version 0.1.3. Queen's University) in the training cohort (Guangzhou training cohort, n = 843). We used the neural network DeepSurv to analyze the pathological microscopic features (DSPMF) and then classified patients into high-risk and low-risk groups through the time-dependent receiver operating characteristic (ROC). The prognosis accuracy of the pathological feature was validated in a validation cohort (n = 212). The primary endpoint was progression-free survival (PFS). RESULTS We found 429 pathological microscopic features in the H&E image. Patients with high-risk scores in the training cohort had shorter 5-year PFS (HR 10.03, 6.06-16.61; P < .0001). The DSPMF (C-index: 0.723) had the higher C-index than the EBV DNA (C-index: 0.612) copies and the N stage (C-index: 0.593). Furthermore, induction chemotherapy (ICT) plus concomitant chemoradiotherapy (CCRT) had better 5-year PFS to those received CCRT (P < .0001) in the high-risk group. CONCLUSION The DSPMF is a reliable prognostic tool for survival risk in patients with NPC and might be able to guide the treatment decision.
Collapse
Affiliation(s)
- Kuiyuan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of nasopharyngeal carcinoma, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| | - Weixiong Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of nasopharyngeal carcinoma, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| | - Mengyun Qiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of nasopharyngeal carcinoma, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| | - Xi Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of nasopharyngeal carcinoma, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| | - Jia Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of Intensive Care Center, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| | - Xiang Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of nasopharyngeal carcinoma, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| | - Xing Lv
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,Department of nasopharyngeal carcinoma, Sun Yat-sen University Cancer Centre, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Nawaz S, Trahearn NA, Heindl A, Banerjee S, Maley CC, Sottoriva A, Yuan Y. Analysis of tumour ecological balance reveals resource-dependent adaptive strategies of ovarian cancer. EBioMedicine 2019; 48:224-235. [PMID: 31648981 PMCID: PMC6838425 DOI: 10.1016/j.ebiom.2019.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/02/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Despite treatment advances, there remains a significant risk of recurrence in ovarian cancer, at which stage it is usually incurable. Consequently, there is a clear need for improved patient stratification. However, at present clinical prognosticators remain largely unchanged due to the lack of reproducible methods to identify high-risk patients. METHODS In high-grade serous ovarian cancer patients with advanced disease, we spatially define a tumour ecological balance of stromal resource and immune hazard using high-throughput image and spatial analysis of routine histology slides. On this basis an EcoScore is developed to classify tumours by a shift in this balance towards cancer-favouring or inhibiting conditions. FINDINGS The EcoScore provides prognostic value stronger than, and independent of, known risk factors. Crucially, the clinical relevance of mutational burden and genomic instability differ under different stromal resource conditions, suggesting that the selective advantage of these cancer hallmarks is dependent on the context of stromal spatial structure. Under a high resource condition defined by a high level of geographical intermixing of cancer and stromal cells, selection appears to be driven by point mutations; whereas, in low resource tumours featured with high hypoxia and low cancer-immune co-localization, selection is fuelled by aneuploidy. INTERPRETATION Our study offers empirical evidence that cancer fitness depends on tumour spatial constraints, and presents a biological basis for developing better assessments of tumour adaptive strategies in overcoming ecological constraints including immune surveillance and hypoxia.
Collapse
Affiliation(s)
- Sidra Nawaz
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK; Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Nicholas A Trahearn
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK; Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Andreas Heindl
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK; Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | | | - Carlo C Maley
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK; Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK; Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK; Division of Molecular Pathology, Institute of Cancer Research, London, UK.
| |
Collapse
|
20
|
Lan C, Li J, Huang X, Heindl A, Wang Y, Yan S, Yuan Y. Stromal cell ratio based on automated image analysis as a predictor for platinum-resistant recurrent ovarian cancer. BMC Cancer 2019; 19:159. [PMID: 30777045 PMCID: PMC6380057 DOI: 10.1186/s12885-019-5343-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 02/01/2019] [Indexed: 02/03/2023] Open
Abstract
Background Identifying high-risk patients for platinum resistance is critical for improving clinical management of ovarian cancer. We aimed to use automated image analysis of hematoxylin & eosin (H&E) stained sections to identify the association between microenvironmental composition and platinum-resistant recurrent ovarian cancer. Methods Ninety-one patients with ovarian cancer containing the data of automated image analysis for H&E histological sections were initially reviewed. Results Seventy-one patients with recurrent disease were finally identified. Among 30 patients with high stromal cell ratio, 60% of the patients had platinum-resistant recurrence, which was significantly higher than the rate in patients with low stromal cell ratio (9.80%, P < 0.001). Multivariate logistic regression analysis revealed elevated CA125 level after 3 cycles of chemotherapy (P < 0.001) and high stromal cell ratio (P = 0.002) were the negative predictors of platinum-resistant relapse. The area under the curve (AUC) of receiver operating characteristic (ROC) curves of the models for predicting platinum-resistant recurrence with stromal cell ratio, normalization of CA125 level, and the combination of two parameters were 0.78, 0.79, and 0.89 respectively. Conclusions Our results demonstrated stromal cell ratio based on automated image analysis may be a potential predictor for ovarian cancer patients at high risk of platinum-resistant recurrence, and it could improve the predictive value of model when combined with normalization of CA125 level after 3 cycles of chemotherapy.
Collapse
Affiliation(s)
- C Lan
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - J Li
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - X Huang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - A Heindl
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.,Centre for Molecular Pathology, The Royal Marsden Hospital, London, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Y Wang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - S Yan
- Department of Pathology, Sun Yat-sen University Cancer Centre, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Y Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK. .,Centre for Molecular Pathology, The Royal Marsden Hospital, London, UK. .,Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
| |
Collapse
|
21
|
Heindl A, Khan AM, Rodrigues DN, Eason K, Sadanandam A, Orbegoso C, Punta M, Sottoriva A, Lise S, Banerjee S, Yuan Y. Microenvironmental niche divergence shapes BRCA1-dysregulated ovarian cancer morphological plasticity. Nat Commun 2018. [PMID: 30254278 DOI: 10.1038/s41467-018-06130-3] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
How tumor microenvironmental forces shape plasticity of cancer cell morphology is poorly understood. Here, we conduct automated histology image and spatial statistical analyses in 514 high grade serous ovarian samples to define cancer morphological diversification within the spatial context of the microenvironment. Tumor spatial zones, where cancer cell nuclei diversify in shape, are mapped in each tumor. Integration of this spatially explicit analysis with omics and clinical data reveals a relationship between morphological diversification and the dysregulation of DNA repair, loss of nuclear integrity, and increased disease mortality. Within the Immunoreactive subtype, spatial analysis further reveals significantly lower lymphocytic infiltration within diversified zones compared with other tumor zones, suggesting that even immune-hot tumors contain cells capable of immune escape. Our findings support a model whereby a subpopulation of morphologically plastic cancer cells with dysregulated DNA repair promotes ovarian cancer progression through positive selection by immune evasion.
Collapse
Affiliation(s)
- Andreas Heindl
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Adnan Mujahid Khan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Daniel Nava Rodrigues
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Katherine Eason
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.,Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Cecilia Orbegoso
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Marco Punta
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Stefano Lise
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK.,Division of Clinical Studies, the Institute of Cancer Research, London, UK, SM2 5NG
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK. .,Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| |
Collapse
|
22
|
Heindl A, Khan AM, Rodrigues DN, Eason K, Sadanandam A, Orbegoso C, Punta M, Sottoriva A, Lise S, Banerjee S, Yuan Y. Microenvironmental niche divergence shapes BRCA1-dysregulated ovarian cancer morphological plasticity. Nat Commun 2018; 9:3917. [PMID: 30254278 PMCID: PMC6156340 DOI: 10.1038/s41467-018-06130-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/15/2018] [Indexed: 12/22/2022] Open
Abstract
How tumor microenvironmental forces shape plasticity of cancer cell morphology is poorly understood. Here, we conduct automated histology image and spatial statistical analyses in 514 high grade serous ovarian samples to define cancer morphological diversification within the spatial context of the microenvironment. Tumor spatial zones, where cancer cell nuclei diversify in shape, are mapped in each tumor. Integration of this spatially explicit analysis with omics and clinical data reveals a relationship between morphological diversification and the dysregulation of DNA repair, loss of nuclear integrity, and increased disease mortality. Within the Immunoreactive subtype, spatial analysis further reveals significantly lower lymphocytic infiltration within diversified zones compared with other tumor zones, suggesting that even immune-hot tumors contain cells capable of immune escape. Our findings support a model whereby a subpopulation of morphologically plastic cancer cells with dysregulated DNA repair promotes ovarian cancer progression through positive selection by immune evasion.
Collapse
Affiliation(s)
- Andreas Heindl
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Adnan Mujahid Khan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Daniel Nava Rodrigues
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Katherine Eason
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK
- Centre for Molecular Pathology, Royal Marsden Hospital, London, SM2 5NG, UK
| | - Cecilia Orbegoso
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Marco Punta
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Stefano Lise
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
- Division of Clinical Studies, the Institute of Cancer Research, London, UK, SM2 5NG
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK.
- Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| |
Collapse
|
23
|
Microenvironmental niche divergence shapes BRCA1-dysregulated ovarian cancer morphological plasticity. Nat Commun 2018. [PMID: 30254278 DOI: 10.1038/s41467-018-06130-3]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
How tumor microenvironmental forces shape plasticity of cancer cell morphology is poorly understood. Here, we conduct automated histology image and spatial statistical analyses in 514 high grade serous ovarian samples to define cancer morphological diversification within the spatial context of the microenvironment. Tumor spatial zones, where cancer cell nuclei diversify in shape, are mapped in each tumor. Integration of this spatially explicit analysis with omics and clinical data reveals a relationship between morphological diversification and the dysregulation of DNA repair, loss of nuclear integrity, and increased disease mortality. Within the Immunoreactive subtype, spatial analysis further reveals significantly lower lymphocytic infiltration within diversified zones compared with other tumor zones, suggesting that even immune-hot tumors contain cells capable of immune escape. Our findings support a model whereby a subpopulation of morphologically plastic cancer cells with dysregulated DNA repair promotes ovarian cancer progression through positive selection by immune evasion.
Collapse
|
24
|
Mlynska A, Povilaityte E, Zemleckaite I, Zilionyte K, Strioga M, Krasko J, Dobrovolskiene N, Peng MW, Intaite B, Pasukoniene V. Platinum sensitivity of ovarian cancer cells does not influence their ability to induce M2-type macrophage polarization. Am J Reprod Immunol 2018; 80:e12996. [PMID: 29904979 DOI: 10.1111/aji.12996] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/23/2018] [Indexed: 12/21/2022] Open
Abstract
PROBLEM Development of platinum resistance in ovarian cancer is mediated by both cancer cells and tumor microenvironment. Activation of epithelial-mesenchymal transition program in cancer cells may lead to enrichment for resistant clones. These processes can be affected by tumor-associated macrophages, a highly plastic population of cells that participate in tumor progression and response to treatment by shaping the microenvironment. We aimed to study how platinum resistance influences the crosstalk between macrophages and ovarian cancer cells. METHOD OF STUDY Using cisplatin-sensitive ovarian cancer cell line A2780, we developed and characterized cisplatin-resistant A2780Cis and cisplatin and doxorubicin co-resistant A2780Dox cell lines. Next, we set up an indirect coculture system with THP-1 cell line-derived M0-type-, M1-type- and M2-type-like polarized macrophages. We monitored the expression of genes associated with cellular stemness, multidrug resistance, and epithelial-mesenchymal transition in cancer cells, and expression profile of M1/M2 markers in macrophages. RESULTS Development of drug resistance in ovarian cancer cell lines was accompanied by increased migration, clonogenicity, and upregulated expression of transcription factors, associated with cellular stemness and epithelial-mesenchymal transition. Upon coculture, we noted that the most relevant changes in gene expression profile occurred in A2780 cells. Moreover, M0- and M1-type macrophages, but not M2-type macrophages, showed significant transcriptional alterations. CONCLUSION Our results provide the evidence for bidirectional interplay between cancer cells and macrophages. Independent of platinum resistance status, ovarian cancer cells polarize macrophages toward M2-like type, whereas macrophages induce epithelial-mesenchymal transition and stemness-related gene expression profile in cisplatin-sensitive, but not cisplatin-resistant cancer cells.
Collapse
Affiliation(s)
- Agata Mlynska
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Egle Povilaityte
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| | - Inga Zemleckaite
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| | - Karolina Zilionyte
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Marius Strioga
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| | - Jan Krasko
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| | | | - Mei-Wen Peng
- Swiss Institute for Experimental Cancer Research, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Birute Intaite
- Department of Oncogynecology, National Cancer Institute, Vilnius, Lithuania
| | - Vita Pasukoniene
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| |
Collapse
|
25
|
Popovici V, Budinská E, Dušek L, Kozubek M, Bosman F. Image-based surrogate biomarkers for molecular subtypes of colorectal cancer. Bioinformatics 2018; 33:2002-2009. [PMID: 28158480 DOI: 10.1093/bioinformatics/btx027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 01/17/2017] [Indexed: 12/15/2022] Open
Abstract
Motivation Whole genome expression profiling of large cohorts of different types of cancer led to the identification of distinct molecular subcategories (subtypes) that may partially explain the observed inter-tumoral heterogeneity. This is also the case of colorectal cancer (CRC) where several such categorizations have been proposed. Despite recent developments, the problem of subtype definition and recognition remains open, one of the causes being the intrinsic heterogeneity of each tumor, which is difficult to estimate from gene expression profiles. However, one of the observations of these studies indicates that there may be links between the dominant tumor morphology characteristics and the molecular subtypes. Benefiting from a large collection of CRC samples, comprising both gene expression and histopathology images, we investigated the possibility of building image-based classifiers able to predict the molecular subtypes. We employed deep convolutional neural networks for extracting local descriptors which were then used for constructing a dictionary-based representation of each tumor sample. A set of support vector machine classifiers were trained to solve different binary decision problems, their combined outputs being used to predict one of the five molecular subtypes. Results A hierarchical decomposition of the multi-class problem was obtained with an overall accuracy of 0.84 (95%CI=0.79-0.88). The predictions from the image-based classifier showed significant prognostic value similar to their molecular counterparts. Contact popovici@iba.muni.cz. Availability and Implementation Source code used for the image analysis is freely available from https://github.com/higex/qpath . Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Vlad Popovici
- Faculty of Medicine, Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Eva Budinská
- Faculty of Science, Research Centre for Toxic Compounds in the Environment, Masaryk University, Brno, Czech Republic
| | - Ladislav Dušek
- Faculty of Medicine, Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Michal Kozubek
- Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Fred Bosman
- University Institute of Pathology, University of Lausanne, Switzerland
| |
Collapse
|
26
|
Heindl A, Lan C, Rodrigues DN, Koelble K, Yuan Y. Similarity and diversity of the tumor microenvironment in multiple metastases: critical implications for overall and progression-free survival of high-grade serous ovarian cancer. Oncotarget 2018; 7:71123-71135. [PMID: 27661102 PMCID: PMC5342067 DOI: 10.18632/oncotarget.12106] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 08/24/2016] [Indexed: 12/29/2022] Open
Abstract
The tumor microenvironment is pivotal in influencing cancer progression and metastasis. Different cells co-exist with high spatial diversity within a patient, yet their combinatorial effects are poorly understood. We investigate the similarity of the tumor microenvironment of 192 local metastatic lesions in 61 ovarian cancer patients. An ecologically inspired measure of microenvironmental diversity derived from multiple metastasis sites is correlated with clinicopathological characteristics and prognostic outcome. We demonstrate a high accuracy of our automated analysis across multiple sites. A low level of similarity in microenvironmental composition is observed between ovary tumor and corresponding local metastases (stromal ratio r = 0.30, lymphocyte ratio r = 0.37). We identify a new measure of microenvironmental diversity derived from Shannon entropy that is highly predictive of poor overall (p = 0.002, HR = 3.18, 95% CI = 1.51-6.68) and progression-free survival (p = 0.0036, HR = 2.83, 95% CI = 1.41-5.7), independent of and stronger than clinical variables, subtype stratifications based on single cell types alone and number of sites. Although stromal influence in ovary tumors is known to have significant clinical implications, our findings reveal an even stronger impact orchestrated by diverse cell types. Quantitative histology-based measures can further enable objective selection of patients who are in urgent need of new therapeutic strategies such as combinatorial treatments targeting heterogeneous tumor microenvironment.
Collapse
Affiliation(s)
- Andreas Heindl
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.,Centre for Molecular Pathology, Royal Marsden Hospital, London, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Chunyan Lan
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Centre, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | | | - Konrad Koelble
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.,Department of Histopathology, Royal Marsden Hospital, London, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.,Centre for Molecular Pathology, Royal Marsden Hospital, London, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
27
|
Shashni B, Ariyasu S, Takeda R, Suzuki T, Shiina S, Akimoto K, Maeda T, Aikawa N, Abe R, Osaki T, Itoh N, Aoki S. Size-Based Differentiation of Cancer and Normal Cells by a Particle Size Analyzer Assisted by a Cell-Recognition PC Software. Biol Pharm Bull 2018; 41:487-503. [PMID: 29332929 DOI: 10.1248/bpb.b17-00776] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Detection of anomalous cells such as cancer cells from normal blood cells has the potential to contribute greatly to cancer diagnosis and therapy. Conventional methods for the detection of cancer cells are usually tedious and cumbersome. Herein, we report on the use of a particle size analyzer for the convenient size-based differentiation of cancer cells from normal cells. Measurements made using a particle size analyzer revealed that size parameters for cancer cells are significantly greater (e.g., inner diameter and width) than the corresponding values for normal cells (white blood cells (WBC), lymphocytes and splenocytes), with no significant difference in shape parameters (e.g., circularity and convexity). The inner diameter of many cancer cell lines is greater than 10 µm, in contrast to normal cells. For the detection of WBC having similar size to that of cancer cells, we developed a PC software "Cancer Cell Finder" that differentiates them from cancer cells based on brightness stationary points on a cell surface. Furthermore, the aforementioned method was validated for cancer cell/clusters detection in spiked mouse blood samples (a B16 melanoma mouse xenograft model) and circulating tumor cell cluster-like particles in the cat and dog (diagnosed with cancer) blood samples. These results provide insights into the possible applicability of the use of a particle size analyzer in conjunction with PC software for the convenient detection of cancer cells in experimental and clinical samples for theranostics.
Collapse
Affiliation(s)
- Babita Shashni
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Shinya Ariyasu
- Center for Technologies Against Cancer, Tokyo University of Science
| | - Reisa Takeda
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Toshihiro Suzuki
- Research Institute for Biomedical Sciences, Tokyo University of Science
| | - Shota Shiina
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Kazunori Akimoto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science.,Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science
| | - Takuto Maeda
- Faculty of Industrial Science and Technology, Tokyo University of Science
| | - Naoyuki Aikawa
- Center for Technologies Against Cancer, Tokyo University of Science.,Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science.,Faculty of Industrial Science and Technology, Tokyo University of Science
| | - Ryo Abe
- Center for Technologies Against Cancer, Tokyo University of Science.,Research Institute for Biomedical Sciences, Tokyo University of Science.,Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science
| | - Tomohiro Osaki
- Laboratory of Veterinary Surgery, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University
| | - Norihiko Itoh
- Laboratory of Veterinary Surgery, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science.,Center for Technologies Against Cancer, Tokyo University of Science.,Division of Medical Science-Engineering Corporation, Research Institute for Science and Technology, Tokyo University of Science
| |
Collapse
|
28
|
Ing N, Huang F, Conley A, You S, Ma Z, Klimov S, Ohe C, Yuan X, Amin MB, Figlin R, Gertych A, Knudsen BS. A novel machine learning approach reveals latent vascular phenotypes predictive of renal cancer outcome. Sci Rep 2017; 7:13190. [PMID: 29038551 PMCID: PMC5643431 DOI: 10.1038/s41598-017-13196-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/19/2017] [Indexed: 12/19/2022] Open
Abstract
Gene expression signatures are commonly used as predictive biomarkers, but do not capture structural features within the tissue architecture. Here we apply a 2-step machine learning framework for quantitative imaging of tumor vasculature to derive a spatially informed, prognostic gene signature. The trained algorithms classify endothelial cells and generate a vascular area mask (VAM) in H&E micrographs of clear cell renal cell carcinoma (ccRCC) cases from The Cancer Genome Atlas (TCGA). Quantification of VAMs led to the discovery of 9 vascular features (9VF) that predicted disease-free-survival in a discovery cohort (n = 64, HR = 2.3). Correlation analysis and information gain identified a 14 gene expression signature related to the 9VF's. Two generalized linear models with elastic net regularization (14VF and 14GT), based on the 14 genes, separated independent cohorts of up to 301 cases into good and poor disease-free survival groups (14VF HR = 2.4, 14GT HR = 3.33). For the first time, we successfully applied digital image analysis and targeted machine learning to develop prognostic, morphology-based, gene expression signatures from the vascular architecture. This novel morphogenomic approach has the potential to improve previous methods for biomarker development.
Collapse
Affiliation(s)
- Nathan Ing
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Fangjin Huang
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew Conley
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Zhaoxuan Ma
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Sergey Klimov
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Chisato Ohe
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaopu Yuan
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Mahul B Amin
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Robert Figlin
- Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Arkadiusz Gertych
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, CA, USA.
| | - Beatrice S Knudsen
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
29
|
Tennill TA, Gross ME, Frieboes HB. Automated analysis of co-localized protein expression in histologic sections of prostate cancer. PLoS One 2017; 12:e0178362. [PMID: 28552967 PMCID: PMC5446169 DOI: 10.1371/journal.pone.0178362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/11/2017] [Indexed: 12/13/2022] Open
Abstract
An automated approach based on routinely-processed, whole-slide immunohistochemistry (IHC) was implemented to study co-localized protein expression in tissue samples. Expression of two markers was chosen to represent stromal (CD31) and epithelial (Ki-67) compartments in prostate cancer. IHC was performed on whole-slide sections representing low-, intermediate-, and high-grade disease from 15 patients. The automated workflow was developed using a training set of regions-of-interest in sequential tissue sections. Protein expression was studied on digital representations of IHC images across entire slides representing formalin-fixed paraffin embedded blocks. Using the training-set, the known association between Ki-67 and Gleason grade was confirmed. CD31 expression was more heterogeneous across samples and remained invariant with grade in this cohort. Interestingly, the Ki-67/CD31 ratio was significantly increased in high (Gleason ≥ 8) versus low/intermediate (Gleason ≤7) samples when assessed in the training-set and the whole-tissue block images. Further, the feasibility of the automated approach to process Tissue Microarray (TMA) samples in high throughput was evaluated. This work establishes an initial framework for automated analysis of co-localized protein expression and distribution in high-resolution digital microscopy images based on standard IHC techniques. Applied to a larger sample population, the approach may help to elucidate the biologic basis for the Gleason grade, which is the strongest, single factor distinguishing clinically aggressive from indolent prostate cancer.
Collapse
Affiliation(s)
- Thomas A. Tennill
- Department of Bioengineering, University of Louisville, Louisville, KY, United States of America
| | - Mitchell E. Gross
- Lawrence J. Elliston Institute for Transformational Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States of America
| |
Collapse
|
30
|
Vandenberghe ME, Scott MLJ, Scorer PW, Söderberg M, Balcerzak D, Barker C. Relevance of deep learning to facilitate the diagnosis of HER2 status in breast cancer. Sci Rep 2017; 7:45938. [PMID: 28378829 PMCID: PMC5380996 DOI: 10.1038/srep45938] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/06/2017] [Indexed: 11/10/2022] Open
Abstract
Tissue biomarker scoring by pathologists is central to defining the appropriate therapy for patients with cancer. Yet, inter-pathologist variability in the interpretation of ambiguous cases can affect diagnostic accuracy. Modern artificial intelligence methods such as deep learning have the potential to supplement pathologist expertise to ensure constant diagnostic accuracy. We developed a computational approach based on deep learning that automatically scores HER2, a biomarker that defines patient eligibility for anti-HER2 targeted therapies in breast cancer. In a cohort of 71 breast tumour resection samples, automated scoring showed a concordance of 83% with a pathologist. The twelve discordant cases were then independently reviewed, leading to a modification of diagnosis from initial pathologist assessment for eight cases. Diagnostic discordance was found to be largely caused by perceptual differences in assessing HER2 expression due to high HER2 staining heterogeneity. This study provides evidence that deep learning aided diagnosis can facilitate clinical decision making in breast cancer by identifying cases at high risk of misdiagnosis.
Collapse
Affiliation(s)
- Michel E. Vandenberghe
- Personalised Healthcare & Biomarkers, IMED Biotech Unit, AstraZeneca, HODGKIN, C/o B310 Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, United Kingdom
| | - Marietta L. J. Scott
- Personalised Healthcare & Biomarkers, IMED Biotech Unit, AstraZeneca, HODGKIN, C/o B310 Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, United Kingdom
| | - Paul W. Scorer
- Personalised Healthcare & Biomarkers, IMED Biotech Unit, AstraZeneca, HODGKIN, C/o B310 Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, United Kingdom
| | - Magnus Söderberg
- Pathology, Drug Safety & Metabolism, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, 431 50 Mölndal, Sweden
| | - Denis Balcerzak
- Personalised Healthcare & Biomarkers, IMED Biotech Unit, AstraZeneca, HODGKIN, C/o B310 Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, United Kingdom
| | - Craig Barker
- Personalised Healthcare & Biomarkers, IMED Biotech Unit, AstraZeneca, HODGKIN, C/o B310 Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, United Kingdom
| |
Collapse
|
31
|
Simmons A, Burrage PM, Nicolau DV, Lakhani SR, Burrage K. Environmental factors in breast cancer invasion: a mathematical modelling review. Pathology 2017; 49:172-180. [PMID: 28081961 DOI: 10.1016/j.pathol.2016.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/07/2016] [Accepted: 11/13/2016] [Indexed: 12/17/2022]
Abstract
This review presents a brief overview of breast cancer, focussing on its heterogeneity and the role of mathematical modelling and simulation in teasing apart the underlying biophysical processes. Following a brief overview of the main known pathophysiological features of ductal carcinoma, attention is paid to differential equation-based models (both deterministic and stochastic), agent-based modelling, multi-scale modelling, lattice-based models and image-driven modelling. A number of vignettes are presented where these modelling approaches have elucidated novel aspects of breast cancer dynamics, and we conclude by offering some perspectives on the role mathematical modelling can play in understanding breast cancer development, invasion and treatment therapies.
Collapse
Affiliation(s)
- Alex Simmons
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia
| | - Pamela M Burrage
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia
| | - Dan V Nicolau
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia; Mathematical Institute, University of Oxford, Oxford, United Kingdom; Molecular Sense Ltd, Oxford, United Kingdom
| | - Sunil R Lakhani
- The University of Queensland, Centre for Clinical Research and School of Medicine and Pathology Queensland, The Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - Kevin Burrage
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia; Department of Computer Science, University of Oxford, United Kingdom.
| |
Collapse
|
32
|
Griffin J, Treanor D. Digital pathology in clinical use: where are we now and what is holding us back? Histopathology 2016; 70:134-145. [DOI: 10.1111/his.12993] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Jon Griffin
- Sheffield NHS Foundation Trust; Sheffield UK
| | | |
Collapse
|
33
|
Zhong C, Han J, Borowsky A, Parvin B, Wang Y, Chang H. When machine vision meets histology: A comparative evaluation of model architecture for classification of histology sections. Med Image Anal 2016; 35:530-543. [PMID: 27644083 DOI: 10.1016/j.media.2016.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 08/12/2016] [Accepted: 08/26/2016] [Indexed: 12/18/2022]
Abstract
Classification of histology sections in large cohorts, in terms of distinct regions of microanatomy (e.g., stromal) and histopathology (e.g., tumor, necrosis), enables the quantification of tumor composition, and the construction of predictive models of genomics and clinical outcome. To tackle the large technical variations and biological heterogeneities, which are intrinsic in large cohorts, emerging systems utilize either prior knowledge from pathologists or unsupervised feature learning for invariant representation of the underlying properties in the data. However, to a large degree, the architecture for tissue histology classification remains unexplored and requires urgent systematical investigation. This paper is the first attempt to provide insights into three fundamental questions in tissue histology classification: I. Is unsupervised feature learning preferable to human engineered features? II. Does cellular saliency help? III. Does the sparse feature encoder contribute to recognition? We show that (a) in I, both Cellular Morphometric Feature and features from unsupervised feature learning lead to superior performance when compared to SIFT and [Color, Texture]; (b) in II, cellular saliency incorporation impairs the performance for systems built upon pixel-/patch-level features; and (c) in III, the effect of the sparse feature encoder is correlated with the robustness of features, and the performance can be consistently improved by the multi-stage extension of systems built upon both Cellular Morphmetric Feature and features from unsupervised feature learning. These insights are validated with two cohorts of Glioblastoma Multiforme (GBM) and Kidney Clear Cell Carcinoma (KIRC).
Collapse
Affiliation(s)
- Cheng Zhong
- Lawrence Berkeley National Laboratory, Berkeley CA USA
| | - Ju Han
- Lawrence Berkeley National Laboratory, Berkeley CA USA
| | - Alexander Borowsky
- Center for Comparative Medicine, University of California, Davis,CA, USA
| | - Bahram Parvin
- Department of Electrical and Biomedical Engineering, University of Nevada, Reno, NV USA
| | - Yunfu Wang
- Lawrence Berkeley National Laboratory, Berkeley CA USA; Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hang Chang
- Lawrence Berkeley National Laboratory, Berkeley CA USA.
| |
Collapse
|
34
|
Kather JN, Weis CA, Bianconi F, Melchers SM, Schad LR, Gaiser T, Marx A, Zöllner FG. Multi-class texture analysis in colorectal cancer histology. Sci Rep 2016; 6:27988. [PMID: 27306927 PMCID: PMC4910082 DOI: 10.1038/srep27988] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/25/2016] [Indexed: 02/08/2023] Open
Abstract
Automatic recognition of different tissue types in histological images is an essential part in the digital pathology toolbox. Texture analysis is commonly used to address this problem; mainly in the context of estimating the tumour/stroma ratio on histological samples. However, although histological images typically contain more than two tissue types, only few studies have addressed the multi-class problem. For colorectal cancer, one of the most prevalent tumour types, there are in fact no published results on multiclass texture separation. In this paper we present a new dataset of 5,000 histological images of human colorectal cancer including eight different types of tissue. We used this set to assess the classification performance of a wide range of texture descriptors and classifiers. As a result, we found an optimal classification strategy that markedly outperformed traditional methods, improving the state of the art for tumour-stroma separation from 96.9% to 98.6% accuracy and setting a new standard for multiclass tissue separation (87.4% accuracy for eight classes). We make our dataset of histological images publicly available under a Creative Commons license and encourage other researchers to use it as a benchmark for their studies.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Institute of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Susanne M. Melchers
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Lothar R. Schad
- Institute of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Timo Gaiser
- Institute of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Frank Gerrit Zöllner
- Institute of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|