1
|
Christyani G, Carswell M, Qin S, Kim W. An Overview of Advances in Rare Cancer Diagnosis and Treatment. Int J Mol Sci 2024; 25:1201. [PMID: 38256274 PMCID: PMC10815984 DOI: 10.3390/ijms25021201] [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: 12/01/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer stands as the leading global cause of mortality, with rare cancer comprising 230 distinct subtypes characterized by infrequent incidence. Despite the inherent challenges in addressing the diagnosis and treatment of rare cancers due to their low occurrence rates, several biomedical breakthroughs have led to significant advancement in both areas. This review provides a comprehensive overview of state-of-the-art diagnostic techniques that encompass new-generation sequencing and multi-omics, coupled with the integration of artificial intelligence and machine learning, that have revolutionized rare cancer diagnosis. In addition, this review highlights the latest innovations in rare cancer therapeutic options, comprising immunotherapy, targeted therapy, transplantation, and drug combination therapy, that have undergone clinical trials and significantly contribute to the tumor remission and overall survival of rare cancer patients. In this review, we summarize recent breakthroughs and insights in the understanding of rare cancer pathophysiology, diagnosis, and therapeutic modalities, as well as the challenges faced in the development of rare cancer diagnosis data interpretation and drug development.
Collapse
Affiliation(s)
| | | | - Sisi Qin
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea; (G.C.); (M.C.)
| | - Wootae Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea; (G.C.); (M.C.)
| |
Collapse
|
2
|
Zhang Z, Wu B, Shao Y, Chen Y, Wang D. A systematic review verified by bioinformatic analysis based on TCGA reveals week prognosis power of CAIX in renal cancer. PLoS One 2022; 17:e0278556. [PMID: 36542612 PMCID: PMC9770376 DOI: 10.1371/journal.pone.0278556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Carbonic anhydrase IX (CAIX) protein has been correlated with progression and survival in patients with some tumors such as head and neck carcinoma. But renal cell carcinoma is an exception. The prognostic value of CAIX in RCC used to be associated with patients' survival according to published works. This study aimed to rectify the former conclusion. METHODS This study was registered in PROSPERO (CRD42020160181). A literature search of the PubMed, Embase, Cochrane library and Web of Science databases was performed to retrieve original studies until April of 2022. Twenty-seven studies, including a total of 5462 patients with renal cell carcinoma, were reviewed. Standard meta-analysis methods were used to evaluate the prognostic impact of CAIX expression on patient prognosis. The hazard ratio and its 95% confidence interval were recorded for the relationship between CAIX expression and survival, and the data were analyzed using Stata 11.0. Then we verify the meta-analysis resort to bioinformatics (TCGA). RESULTS Our initial search resulted in 908 articles in total. From PubMed, Embase, Web of Science electronic and Cochrane library databases, 493, 318 and 97 potentially relevant articles were discovered, respectively. We took the analysis between CA9 and disease-specific survival (HR = 1.18, 95% CI: 0.82-1.70, I2 = 79.3%, P<0.05), a subgroup then was performed to enhance the result (HR = 1.63, 95%CI: 1.30-2.03, I2 = 26.3%, P = 0.228); overall survival was also parallel with the former (HR = 1.13, 95%CI: 0.82-1.56, I2 = 79.8%, P<0.05), then a subgroup also be performed (HR = 0.90, 95%CI:0.75-1.07, I2 = 23.1%, P = 0.246) to verify the result; the analysis between CAIX and progression-free survival got the similar result (HR = 1.73, 95%CI:0.97-3.09, I2 = 82.4%, P<0.05), we also verify the result by subgroup analysis (HR = 1.04, 95%CI:0.79-1.36, I2 = 0.0%, P = 0.465); at last the relationship between CAIX and recurrence-free survival got the same result, too (HR = 0.99, 95%CI: 0.95-1.02, I2 = 57.8%, P = 0.050), the subgroup's result was also parallel with the former (HR = 1.01, 95%CI: 0.91-1.03, I2 = 0.00%, P = 0.704). To validate our meta-analysis, we took a bioinformatic analysis based on TCGA database, survival curve between low and high CAIX expression in four endpoints (DSS, OS, PFI, DFI) have corresponding P value (DSS:P = 0.23, OS:P = 0.77, PFI:P = 0.25, DFI:P = 0.78). CONCLUSIONS CAIX expression in patients with RCC is an exception to predict tumor survival. Both low CAIX expression and high expression are not associated with survivals in RCC patients.
Collapse
Affiliation(s)
- Zikuan Zhang
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Bo Wu
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Yuan Shao
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Yongquan Chen
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Dongwen Wang
- Basic Medicine of Shanxi Medical University, Taiyuan, China
- * E-mail:
| |
Collapse
|
3
|
Sundaram S, Kim EN, Jones GM, Sivagnanam S, Tripathi M, Miremadi A, Di Pietro M, Coussens LM, Fitzgerald RC, Chang YH, Zhuang L. Deciphering the Immune Complexity in Esophageal Adenocarcinoma and Pre-Cancerous Lesions With Sequential Multiplex Immunohistochemistry and Sparse Subspace Clustering Approach. Front Immunol 2022; 13:874255. [PMID: 35663986 PMCID: PMC9161782 DOI: 10.3389/fimmu.2022.874255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/19/2022] [Indexed: 02/05/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) develops from a chronic inflammatory environment across four stages: intestinal metaplasia, known as Barrett's esophagus, low- and high-grade dysplasia, and adenocarcinoma. Although the genomic characteristics of this progression have been well defined via large-scale DNA sequencing, the dynamics of various immune cell subsets and their spatial interactions in their tumor microenvironment remain unclear. Here, we applied a sequential multiplex immunohistochemistry (mIHC) platform with computational image analysis pipelines that allow for the detection of 10 biomarkers in one formalin-fixed paraffin-embedded (FFPE) tissue section. Using this platform and quantitative image analytics, we studied changes in the immune landscape during disease progression based on 40 normal and diseased areas from endoscopic mucosal resection specimens of chemotherapy treatment- naïve patients, including normal esophagus, metaplasia, low- and high-grade dysplasia, and adenocarcinoma. The results revealed a steady increase of FOXP3+ T regulatory cells and a CD163+ myelomonocytic cell subset. In parallel to the manual gating strategy applied for cell phenotyping, we also adopted a sparse subspace clustering (SSC) algorithm allowing the automated cell phenotyping of mIHC-based single-cell data. The algorithm successfully identified comparable cell types, along with significantly enriched FOXP3 T regulatory cells and CD163+ myelomonocytic cells as found in manual gating. In addition, SCC identified a new CSF1R+CD1C+ myeloid lineage, which not only was previously unknown in this disease but also increases with advancing disease stages. This study revealed immune dynamics in EAC progression and highlighted the potential application of a new multiplex imaging platform, combined with computational image analysis on routine clinical FFPE sections, to investigate complex immune populations in tumor ecosystems.
Collapse
Affiliation(s)
- Srinand Sundaram
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Eun Na Kim
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Georgina M. Jones
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Shamilene Sivagnanam
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, United States
| | - Monika Tripathi
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ahmad Miremadi
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Massimiliano Di Pietro
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Lisa M. Coussens
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Rebecca C. Fitzgerald
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Lizhe Zhuang
- Medical Research Council (MRC) Cancer Unit, Hutchison-Medical Research Council (MRC) Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Caligola S, De Sanctis F, Canè S, Ugel S. Breaking the Immune Complexity of the Tumor Microenvironment Using Single-Cell Technologies. Front Genet 2022; 13:867880. [PMID: 35651929 PMCID: PMC9149246 DOI: 10.3389/fgene.2022.867880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/27/2022] [Indexed: 12/31/2022] Open
Abstract
Tumors are not a simple aggregate of transformed cells but rather a complicated ecosystem containing various components, including infiltrating immune cells, tumor-related stromal cells, endothelial cells, soluble factors, and extracellular matrix proteins. Profiling the immune contexture of this intricate framework is now mandatory to develop more effective cancer therapies and precise immunotherapeutic approaches by identifying exact targets or predictive biomarkers, respectively. Conventional technologies are limited in reaching this goal because they lack high resolution. Recent developments in single-cell technologies, such as single-cell RNA transcriptomics, mass cytometry, and multiparameter immunofluorescence, have revolutionized the cancer immunology field, capturing the heterogeneity of tumor-infiltrating immune cells and the dynamic complexity of tenets that regulate cell networks in the tumor microenvironment. In this review, we describe some of the current single-cell technologies and computational techniques applied for immune-profiling the cancer landscape and discuss future directions of how integrating multi-omics data can guide a new "precision oncology" advancement.
Collapse
Affiliation(s)
| | | | | | - Stefano Ugel
- Immunology Section, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
5
|
Immune cell topography predicts response to PD-1 blockade in cutaneous T cell lymphoma. Nat Commun 2021; 12:6726. [PMID: 34795254 PMCID: PMC8602403 DOI: 10.1038/s41467-021-26974-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023] Open
Abstract
Cutaneous T cell lymphomas (CTCL) are rare but aggressive cancers without effective treatments. While a subset of patients derive benefit from PD-1 blockade, there is a critically unmet need for predictive biomarkers of response. Herein, we perform CODEX multiplexed tissue imaging and RNA sequencing on 70 tumor regions from 14 advanced CTCL patients enrolled in a pembrolizumab clinical trial (NCT02243579). We find no differences in the frequencies of immune or tumor cells between responders and non-responders. Instead, we identify topographical differences between effector PD-1+ CD4+ T cells, tumor cells, and immunosuppressive Tregs, from which we derive a spatial biomarker, termed the SpatialScore, that correlates strongly with pembrolizumab response in CTCL. The SpatialScore coincides with differences in the functional immune state of the tumor microenvironment, T cell function, and tumor cell-specific chemokine recruitment and is validated using a simplified, clinically accessible tissue imaging platform. Collectively, these results provide a paradigm for investigating the spatial balance of effector and suppressive T cell activity and broadly leveraging this biomarker approach to inform the clinical use of immunotherapies.
Collapse
|
6
|
Chong PY, Iqbal J, Yeong J, Aw TC, Chan KS, Chui P. Immune Response in Myocardial Injury: In Situ Hybridization and Immunohistochemistry Techniques for SARS-CoV-2 Detection in COVID-19 Autopsies. Front Mol Biosci 2021; 8:658932. [PMID: 34765640 PMCID: PMC8576174 DOI: 10.3389/fmolb.2021.658932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 09/28/2021] [Indexed: 01/18/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) is caused by the newly discovered coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While the lung remains the primary target site of COVID-19 injury, damage to myocardium, and other organs also contribute to the morbidity and mortality of this disease. There is also increasing demand to visualize viral components within tissue specimens. Here we discuss the cardiac autopsy findings of 12 intensive care unit (ICU) naïve and PCR-positive COVID-19 cases using a combination of histological, Immunohistochemical/immunofluorescent and molecular techniques. We performed SARS-CoV-2 qRT-PCR on fresh tissue from all cases; RNA-ISH and IHC for SARS-CoV-2 were performed on selected cases using FFPE tissue from heart. Eight of these patients also had positive post-mortem serology for SARS-CoV-2. Histopathologic changes in the coronary vessels and inflammation of the myocardium as well as in the endocardium were documented which support the reports of a cardiac component to the viral infection. As in the pulmonary reports, widespread platelet and fibrin thrombi were also identified in the cardiac tissue. In keeping with vaccine-induced activation of virus-specific CD4+ and CD8+ T cells, and release of cytokines such as interferon-gamma (IFNγ), we observed similar immune cellular distribution and cytokines in these patients. Immunohistochemical and immunofluorescent localisation for the viral Spike (S-protein) protein and the nucleocapsid protein (NP) were performed; presence of these aggregates may possibly contribute to cardiac ischemia and even remodelling.
Collapse
Affiliation(s)
- Pek Yoon Chong
- Department of Pathology, Sengkang General Hospital, Singapore, Singapore
| | - Jabed Iqbal
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joe Yeong
- Institute of Molecular and Cell Biology, A-STAR, Singapore, Singapore
| | - Tar Choon Aw
- Department of Pathology, Sengkang General Hospital, Singapore, Singapore
| | - Kian Sing Chan
- Department of Molecular Pathology, Singapore General Hospital, Singapore, Singapore
| | - Paul Chui
- Health Science Authority, Singapore, Singapore
| |
Collapse
|
7
|
Wang J, Browne L, Slapetova I, Shang F, Lee K, Lynch J, Beretov J, Whan R, Graham PH, Millar EKA. Multiplexed immunofluorescence identifies high stromal CD68 +PD-L1 + macrophages as a predictor of improved survival in triple negative breast cancer. Sci Rep 2021; 11:21608. [PMID: 34732817 PMCID: PMC8566595 DOI: 10.1038/s41598-021-01116-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) comprises 10-15% of all breast cancers and has a poor prognosis with a high risk of recurrence within 5 years. PD-L1 is an important biomarker for patient selection for immunotherapy but its cellular expression and co-localization within the tumour immune microenvironment and associated prognostic value is not well defined. We aimed to characterise the phenotypes of immune cells expressing PD-L1 and determine their association with overall survival (OS) and breast cancer-specific survival (BCSS). Using tissue microarrays from a retrospective cohort of TNBC patients from St George Hospital, Sydney (n = 244), multiplexed immunofluorescence (mIF) was used to assess staining for CD3, CD8, CD20, CD68, PD-1, PD-L1, FOXP3 and pan-cytokeratin on the Vectra Polaris™ platform and analysed using QuPath. Cox multivariate analyses showed high CD68+PD-L1+ stromal cell counts were associated with improved prognosis for OS (HR 0.56, 95% CI 0.33-0.95, p = 0.030) and BCSS (HR 0.47, 95% CI 0.25-0.88, p = 0.018) in the whole cohort and in patients receiving chemotherapy, improving incrementally upon the predictive value of PD-L1+ alone for BCSS. These data suggest that CD68+PD-L1+ status can provide clinically useful prognostic information to identify sub-groups of patients with good or poor prognosis and guide treatment decisions in TNBC.
Collapse
Affiliation(s)
- James Wang
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
| | - Lois Browne
- Cancer Care Centre, St George Hospital, Kogarah, Australia
| | - Iveta Slapetova
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Kensington, Australia
| | - Fei Shang
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Kensington, Australia
| | - Kirsty Lee
- Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jodi Lynch
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
- Cancer Care Centre, St George Hospital, Kogarah, Australia
| | - Julia Beretov
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- Department of Anatomical Pathology, New South Wales Health Pathology, St George Hospital, Kogarah, Australia
| | - Renee Whan
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Kensington, Australia
| | - Peter H Graham
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
- Cancer Care Centre, St George Hospital, Kogarah, Australia
| | - Ewan K A Millar
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia.
- Department of Anatomical Pathology, New South Wales Health Pathology, St George Hospital, Kogarah, Australia.
- Faculty of Medicine and Health Sciences, Western Sydney University, Campbelltown, Australia.
- University of Technology, Sydney, Australia.
| |
Collapse
|
8
|
Challenges and Opportunities in the Statistical Analysis of Multiplex Immunofluorescence Data. Cancers (Basel) 2021; 13:cancers13123031. [PMID: 34204319 PMCID: PMC8233801 DOI: 10.3390/cancers13123031] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Immune modulation is considered a hallmark of cancer initiation and progression, and has offered promising opportunities for therapeutic manipulation. Multiplex immunofluorescence (mIF) technology has enabled the tumor immune microenvironment (TIME) to be studied at an increased scale, in terms of both the number of markers and the number of samples. Another benefit of mIF technology is the ability to measure not only the abundance but also the spatial location of multiple cells types within a tissue sample simultaneously, allowing for assessment of the co-localization of different types of immune markers. Thus, the use of mIF technologies have enable researchers to characterize patient, clinical, and tumor characteristics in the hope of identifying patients whom might benefit from immunotherapy treatments. In this review we outline some of the challenges and opportunities in the statistical analyses of mIF data to study the TIME. Abstract Immune modulation is considered a hallmark of cancer initiation and progression. The recent development of immunotherapies has ushered in a new era of cancer treatment. These therapeutics have led to revolutionary breakthroughs; however, the efficacy of immunotherapy has been modest and is often restricted to a subset of patients. Hence, identification of which cancer patients will benefit from immunotherapy is essential. Multiplex immunofluorescence (mIF) microscopy allows for the assessment and visualization of the tumor immune microenvironment (TIME). The data output following image and machine learning analyses for cell segmenting and phenotyping consists of the following information for each tumor sample: the number of positive cells for each marker and phenotype(s) of interest, number of total cells, percent of positive cells for each marker, and spatial locations for all measured cells. There are many challenges in the analysis of mIF data, including many tissue samples with zero positive cells or “zero-inflated” data, repeated measurements from multiple TMA cores or tissue slides per subject, and spatial analyses to determine the level of clustering and co-localization between the cell types in the TIME. In this review paper, we will discuss the challenges in the statistical analysis of mIF data and opportunities for further research.
Collapse
|
9
|
Tien TZ, Lee JNLW, Lim JCT, Chen XY, Thike AA, Tan PH, Yeong JPS. Delineating the breast cancer immune microenvironment in the era of multiplex immunohistochemistry/immunofluorescence. Histopathology 2021; 79:139-159. [PMID: 33400265 DOI: 10.1111/his.14328] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common malignancy and the leading cause of cancer death in females worldwide. Treatment is challenging, especially for those who are triple-negative. Increasing evidence suggests that diverse immune populations are present in the breast tumour microenvironment, which opens up avenues for personalised drug targets. Historically, our investigations into the immune constitution of breast tumours have been restricted to analyses of one or two markers at a given time. Recent technological advances have allowed simultaneous labelling of more than 35 markers and detailed profiling of tumour-immune infiltrates at the single-cell level, as well as determining the cellular composition and spatial analysis of the entire tumour architecture. In this review, we describe emerging technologies that have contributed to the field of breast cancer diagnosis, and discuss how to interpret the vast data sets obtained in order to effectively translate them for clinically relevant use.
Collapse
Affiliation(s)
- Tracy Z Tien
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Justina N L W Lee
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jeffrey C T Lim
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xiao-Yang Chen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joe P S Yeong
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
10
|
Petitprez F, Ayadi M, de Reyniès A, Fridman WH, Sautès-Fridman C, Job S. Review of Prognostic Expression Markers for Clear Cell Renal Cell Carcinoma. Front Oncol 2021; 11:643065. [PMID: 33996558 PMCID: PMC8113694 DOI: 10.3389/fonc.2021.643065] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
Context: The number of prognostic markers for clear cell renal cell carcinoma (ccRCC) has been increasing regularly over the last 15 years, without being integrated and compared. Objective: Our goal was to perform a review of prognostic markers for ccRCC to lay the ground for their use in the clinics. Evidence Acquisition: PubMed database was searched to identify RNA and protein markers whose expression level was reported as associated with survival of ccRCC patients. Relevant studies were selected through cross-reading by two readers. Evidence Synthesis: We selected 249 studies reporting an association with prognostic of either single markers or multiple-marker models. Altogether, these studies were based on a total of 341 distinct markers and 13 multiple-marker models. Twenty percent of these markers were involved in four biological pathways altered in ccRCC: cell cycle, angiogenesis, hypoxia, and immune response. The main genes (VHL, PBRM1, BAP1, and SETD2) involved in ccRCC carcinogenesis are not the most relevant for assessing survival. Conclusion: Among single markers, the most validated markers were KI67, BIRC5, TP53, CXCR4, and CA9. Of the multiple-marker models, the most famous model, ClearCode34, has been highly validated on several independent datasets, but its clinical utility has not yet been investigated. Patient Summary: Over the years, the prognosis studies have evolved from single markers to multiple-marker models. Our review highlights the highly validated prognostic markers and multiple-marker models and discusses their clinical utility for better therapeutic care.
Collapse
Affiliation(s)
- Florent Petitprez
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Mira Ayadi
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Wolf H. Fridman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Inflammation, Complément et Cancer, Paris, France
| | - Catherine Sautès-Fridman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Inflammation, Complément et Cancer, Paris, France
| | - Sylvie Job
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| |
Collapse
|
11
|
Chan JY, Lim JQ, Yeong J, Ravi V, Guan P, Boot A, Tay TKY, Selvarajan S, Md Nasir ND, Loh JH, Ong CK, Huang D, Tan J, Li Z, Ng CCY, Tan TT, Masuzawa M, Sung KWK, Farid M, Quek RHH, Tan NC, Teo MCC, Rozen SG, Tan P, Futreal A, Teh BT, Soo KC. Multiomic analysis and immunoprofiling reveal distinct subtypes of human angiosarcoma. J Clin Invest 2021; 130:5833-5846. [PMID: 33016928 DOI: 10.1172/jci139080] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022] Open
Abstract
Angiosarcomas are rare, clinically aggressive tumors with limited treatment options and a dismal prognosis. We analyzed angiosarcomas from 68 patients, integrating information from multiomic sequencing, NanoString immuno-oncology profiling, and multiplex immunohistochemistry and immunofluorescence for tumor-infiltrating immune cells. Through whole-genome sequencing (n = 18), 50% of the cutaneous head and neck angiosarcomas exhibited higher tumor mutation burden (TMB) and UV mutational signatures; others were mutationally quiet and non-UV driven. NanoString profiling revealed 3 distinct patient clusters represented by lack (clusters 1 and 2) or enrichment (cluster 3) of immune-related signaling and immune cells. Neutrophils (CD15+), macrophages (CD68+), cytotoxic T cells (CD8+), Tregs (FOXP3+), and PD-L1+ cells were enriched in cluster 3 relative to clusters 2 and 1. Likewise, tumor inflammation signature (TIS) scores were highest in cluster 3 (7.54 vs. 6.71 vs. 5.75, respectively; P < 0.0001). Head and neck angiosarcomas were predominant in clusters 1 and 3, providing the rationale for checkpoint immunotherapy, especially in the latter subgroup with both high TMB and TIS scores. Cluster 2 was enriched for secondary angiosarcomas and exhibited higher expression of DNMT1, BRD3/4, MYC, HRAS, and PDGFRB, in keeping with the upregulation of epigenetic and oncogenic signaling pathways amenable to targeted therapies. Molecular and immunological dissection of angiosarcomas may provide insights into opportunities for precision medicine.
Collapse
Affiliation(s)
- Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Centre, Singapore
| | - Jing Quan Lim
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Joe Yeong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore.,Institute of Molecular and Cell Biology, Singapore
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peiyong Guan
- Integrated Biostatistics and Bioinformatics Programme
| | - Arnoud Boot
- Integrated Biostatistics and Bioinformatics Programme.,Centre for Computational Biology, and
| | | | | | | | - Jie Hua Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Choon Kiat Ong
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Dachuan Huang
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Jing Tan
- Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore
| | - Zhimei Li
- Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore
| | - Cedric Chuan-Young Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore
| | - Thuan Tong Tan
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Mikio Masuzawa
- Department of Regulation Biochemistry, School of Allied Health Sciences, Kitasato University, Minato City, Tokyo, Japan
| | - Ken Wing-Kin Sung
- Genome Institute of Singapore, A*STAR, Singapore.,School of Computing, National University of Singapore, Singapore
| | - Mohamad Farid
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Centre, Singapore
| | | | - Ngian Chye Tan
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore.,SingHealth Duke-NUS Head and Neck Centre, Singapore
| | | | - Steven George Rozen
- Integrated Biostatistics and Bioinformatics Programme.,Centre for Computational Biology, and.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Patrick Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore.,Genome Institute of Singapore, A*STAR, Singapore
| | - Andrew Futreal
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bin Tean Teh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore.,Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore.,Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Khee Chee Soo
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore.,SingHealth Duke-NUS Head and Neck Centre, Singapore
| |
Collapse
|
12
|
Lam JH, Hong M, Koo SL, Chua CWL, Lim KL, Wee F, Wan WK, Leow WQ, Yeo JG, Tan IBH, Yeong J, Lim TKH, Lim TS. CD30 +OX40 + Treg is associated with improved overall survival in colorectal cancer. Cancer Immunol Immunother 2021; 70:2353-2365. [PMID: 33527196 PMCID: PMC8289785 DOI: 10.1007/s00262-021-02859-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/06/2021] [Indexed: 12/27/2022]
Abstract
Regulatory T cells (Tregs) are often enriched in tumors, where their immunosuppressive function has a key role in tumor persistence and progression. In colorectal cancer (CRC), however, Tregs are frequently associated with an improved clinical outcome. Tumor-infiltrating Tregs have been shown to exhibit a distinct signature comprising the co-stimulatory molecules (OX40, 4-1BB), cytokine receptors (IL1R2, IL21R, CCR8, CD30), and co-inhibitory molecules (PD-L1, TIGIT). Here, we showed by flow cytometry that circulating CD45RO+ Tregs from patients with CRC (n = 25) have elevated CD30 and OX40 expression compared to healthy subjects (n = 14). We identified co-expression of CD30 and OX40 on circulating CD45RO+ Tregs using single-cell images captured by the DEPArray™ system. The frequency of CD30+OX40+CD45RO+ Tregs was significantly higher in CRC patients than in healthy subjects (P < 0.001). Importantly, receiver operating characteristic analysis confirmed that this CD30+OX40+ Treg subset could strongly discriminate between CRC patients and healthy subjects with the highest accuracy of 92.3%, an AUC of 0.92, a sensitivity of 88%, a specificity of 100%, a positive predictive value of 100%, a negative predictive value of 82.35%, and a trade-off value of 3.44%, compared to other Treg subsets. Consistently, multiplex-IHC/IF of tumor-infiltrating Tregs revealed a significant association between high densities of CD30+OX40+ Tregs and improved overall survival; no such association was found for other subsets. These data suggest a potential role for CD30+OX40+ Tregs as a diagnostic or prognostic biomarker in CRC.
Collapse
Affiliation(s)
- Jian Hang Lam
- A. Menarini Biomarkers Singapore Pte Ltd, Singapore, Singapore
| | - Michelle Hong
- A. Menarini Biomarkers Singapore Pte Ltd, Singapore, Singapore
| | - Si-Lin Koo
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | | | - Kah Ling Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Felicia Wee
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Wei Keat Wan
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Wei Qiang Leow
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joo Guan Yeo
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Iain Bee Huat Tan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore, Singapore. .,Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Tony Kiat Hon Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.
| | - Tong Seng Lim
- A. Menarini Biomarkers Singapore Pte Ltd, Singapore, Singapore.
| |
Collapse
|
13
|
Ng HHM, Lee RY, Goh S, Tay ISY, Lim X, Lee B, Chew V, Li H, Tan B, Lim S, Lim JCT, Au B, Loh JJH, Saraf S, Connolly JE, Loh T, Leow WQ, Lee JJX, Toh HC, Malavasi F, Lee SY, Chow P, Newell EW, Choo SP, Tai D, Yeong J, Lim TKH. Immunohistochemical scoring of CD38 in the tumor microenvironment predicts responsiveness to anti-PD-1/PD-L1 immunotherapy in hepatocellular carcinoma. J Immunother Cancer 2020; 8:jitc-2020-000987. [PMID: 32847986 PMCID: PMC7451957 DOI: 10.1136/jitc-2020-000987] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-associated mortality globally. Immune-checkpoint blockade (ICB) is one of the systemic therapy options for HCC. However, response rates remain low, necessitating robust predictive biomarkers. In the present study, we examined the expression of CD38, a molecule involved in the immunosuppressive adenosinergic pathway, on immune cells present in the tumor microenvironment. We then investigated the association between CD38 and ICB treatment outcomes in advanced HCC. Methods Clinically annotated samples from 49 patients with advanced HCC treated with ICB were analyzed for CD38 expression using immunohistochemistry (IHC), multiplex immunohistochemistry/immunofluorescence (mIHC/IF) and multiplex cytokine analysis. Results IHC and mIHC/IF analyses revealed that higher intratumoral CD38+ cell proportion was strongly associated with improved response to ICB. The overall response rates to ICB was significantly higher among patients with high proportion of total CD38+cells compared with patients with low proportion (43.5% vs 3.9%, p=0.019). Higher responses seen among patients with a high intratumoral CD38+cell proportion translated to a longer median progression-free survival (mPFS, 8.21 months vs 1.64 months, p=0.0065) and median overall survival (mOS, 19.06 months vs 9.59 months, p=0.0295). Patients with high CD38+CD68+macrophage density had a better mOS of 34.43 months compared with 9.66 months in patients with low CD38+CD68+ macrophage density. CD38hi macrophages produce more interferon γ (IFN-γ) and related cytokines, which may explain its predictive value when treated with ICB. Conclusions A high proportion of CD38+ cells, determined by IHC, predicts response to ICB and is associated with superior mPFS and OS in advanced HCC.
Collapse
Affiliation(s)
- Harry Ho Man Ng
- Duke-NUS Medical School, Singapore.,Division of Pathology, Singapore General Hospital, Singapore
| | - Ren Yuan Lee
- Division of Pathology, Singapore General Hospital, Singapore.,Nanyang Technological University, Singapore
| | - Siting Goh
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Xinru Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Valerie Chew
- Duke-NUS Medical School, Singapore.,SingHealth Translational Immunology and Inflammation Centre (STIIC), Singapore Health Services Pte Ltd, Singapore
| | - Huihua Li
- Division of Medicine, Singapore General Hospital, Singapore.,Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Benedict Tan
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Sherlly Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Bijin Au
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | | | - Sahil Saraf
- Division of Pathology, Singapore General Hospital, Singapore
| | - John Edward Connolly
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Tracy Loh
- Division of Pathology, Singapore General Hospital, Singapore
| | - Wei Qiang Leow
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Fabio Malavasi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Ser Yee Lee
- Duke-NUS Medical School, Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Pierce Chow
- Duke-NUS Medical School, Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - David Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore .,Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
14
|
Clark DJ, Zhang H. Proteomic approaches for characterizing renal cell carcinoma. Clin Proteomics 2020; 17:28. [PMID: 32742246 PMCID: PMC7391522 DOI: 10.1186/s12014-020-09291-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022] Open
Abstract
Renal cell carcinoma is among the top 15 most commonly diagnosed cancers worldwide, comprising multiple sub-histologies with distinct genomic, proteomic, and clinicopathological features. Proteomic methodologies enable the detection and quantitation of protein profiles associated with the disease state and have been explored to delineate the dysregulated cellular processes associated with renal cell carcinoma. In this review we highlight the reports that employed proteomic technologies to characterize tissue, blood, and urine samples obtained from renal cell carcinoma patients. We describe the proteomic approaches utilized and relate the results of studies in the larger context of renal cell carcinoma biology. Moreover, we discuss some unmet clinical needs and how emerging proteomic approaches can seek to address them. There has been significant progress to characterize the molecular features of renal cell carcinoma; however, despite the large-scale studies that have characterized the genomic and transcriptomic profiles, curative treatments are still elusive. Proteomics facilitates a direct evaluation of the functional modules that drive pathobiology, and the resulting protein profiles would have applications in diagnostics, patient stratification, and identification of novel therapeutic interventions.
Collapse
Affiliation(s)
- David J. Clark
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231 USA
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231 USA
| |
Collapse
|
15
|
Jiang Y, Li W, Huang C, Tian C, Chen Q, Zeng X, Cao Y, Chen Y, Yang Y, Liu H, Bo Y, Luo C, Li Y, Zhang T, Wang R. Preoperative CT Radiomics Predicting the SSIGN Risk Groups in Patients With Clear Cell Renal Cell Carcinoma: Development and Multicenter Validation. Front Oncol 2020; 10:909. [PMID: 32850304 PMCID: PMC7402386 DOI: 10.3389/fonc.2020.00909] [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: 03/05/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
Objective: The stage, size, grade, and necrosis (SSIGN) score can facilitate the assessment of tumor aggressiveness and the personal management for patients with clear cell renal cell carcinoma (ccRCC). However, this score is only available after the postoperative pathological evaluation. The aim of this study was to develop and validate a CT radiomic signature for the preoperative prediction of SSIGN risk groups in patients with ccRCC in multicenters. Methods: In total, 330 patients with ccRCC from three centers were classified into the training, external validation 1, and external validation 2 cohorts. Through consistent analysis and the least absolute shrinkage and selection operator, a radiomic signature was developed to predict the SSIGN low-risk group (scores 0–3) and intermediate- to high-risk group (score ≥ 4). An image feature model was developed according to the independent image features, and a fusion model was constructed integrating the radiomic signature and the independent image features. Furthermore, the predictive performance of the above models for the SSIGN risk groups was evaluated with regard to their discrimination, calibration, and clinical usefulness. Results: A radiomic signature consisting of sixteen relevant features from the nephrographic phase CT images achieved a good calibration (all Hosmer–Lemeshow p > 0.05) and favorable prediction efficacy in the training cohort [area under the curve (AUC): 0.940, 95% confidence interval (CI): 0.884–0.973] and in the external validation cohorts (AUC: 0.876, 95% CI: 0.811–0.942; AUC: 0.928, 95% CI: 0.844–0.975, respectively). The radiomic signature performed better than the image feature model constructed by intra-tumoral vessels (all p < 0.05) and showed similar performance with the fusion model integrating radiomic signature and intra-tumoral vessels (all p > 0.05) in terms of the discrimination in all cohorts. Moreover, the decision curve analysis verified the clinical utility of the radiomic signature in both external cohorts. Conclusion: Radiomic signature could be used as a promising non-invasive tool to predict SSIGN risk groups and to facilitate preoperative clinical decision-making for patients with ccRCC.
Collapse
Affiliation(s)
- Yi Jiang
- Medical College, Guizhou University, Guiyang, China.,Department of Medical Records and Statistics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Wuchao Li
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China.,Guizhou Provincial Key Laboratory of Intelligent Medical Image Analysis and Precision Diagnosis, Guizhou Provincial People's Hospital, Guiyang, China
| | - Chencui Huang
- Research Collaboration Department, R&D Center, Beijing Deepwise & League of PHD Technology Co. Ltd, Beijing, China
| | - Chong Tian
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China.,Guizhou Provincial Key Laboratory of Intelligent Medical Image Analysis and Precision Diagnosis, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qi Chen
- Department of Medical Records and Statistics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xianchun Zeng
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China.,Guizhou Provincial Key Laboratory of Intelligent Medical Image Analysis and Precision Diagnosis, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yin Cao
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yi Chen
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yintong Yang
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Heng Liu
- Department of Radiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yonghua Bo
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chenggong Luo
- Department of Urinary Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yiming Li
- Research Collaboration Department, R&D Center, Beijing Deepwise & League of PHD Technology Co. Ltd, Beijing, China
| | - Tijiang Zhang
- Department of Radiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Rongping Wang
- Medical College, Guizhou University, Guiyang, China.,Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China.,Guizhou Provincial Key Laboratory of Intelligent Medical Image Analysis and Precision Diagnosis, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
16
|
Yamamoto-Fukuda T, Akiyama N, Kojima H. L1CAM-ILK-YAP Mechanotransduction Drives Proliferative Activity of Epithelial Cells in Middle Ear Cholesteatoma. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1667-1679. [PMID: 32360569 DOI: 10.1016/j.ajpath.2020.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/17/2020] [Accepted: 04/15/2020] [Indexed: 02/09/2023]
Abstract
Middle-ear cholesteatoma (cholesteatoma) is a chronic otitis media with an enhanced proliferation of epithelial cells. Negative pressure in the middle ear is thought to be important for the etiology of cholesteatoma. However, the mechanism of cholesteatoma formation remains unclear. Integrin-linked protein kinase (ILK), an important modulator of actin cytoskeletal dynamics, interacts with extracellular matrix and results in the up-regulation of mechanotransduction effector Yes-associated protein (YAP). The L1 cell adhesion molecule (L1CAM) has recently been reported as an activator of the mechanotransduction effectors related to cell proliferation and migration. In this study, we demonstrated a stretch assay for middle-ear cultured cells and performed immunohistochemistry using antibodies against Ilk, Yap, and L1cam. The tympanic membrane was also analyzed within a new middle-ear negative-pressure animal model and human cholesteatoma tissues, using immunohistochemistry with antibodies against ILK, YAP, Ki-67, and L1CAM. The expression of cytoplasmic ILK and nuclear shift of YAP increased in the thickened epithelium of the tympanic membrane under a negative-pressure load and the cholesteatoma. The expression of L1CAM was detected in the stromal cells, which enhanced epithelial cell proliferation depending on ILK signaling events. In conclusion, we demonstrated the possibility that the stromal L1CAM and epithelial ILK-YAP signaling played an important role in epithelial growth under mechanotransduction in cholesteatoma formation.
Collapse
Affiliation(s)
| | - Naotaro Akiyama
- Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Tan WCC, Nerurkar SN, Cai HY, Ng HHM, Wu D, Wee YTF, Lim JCT, Yeong J, Lim TKH. Overview of multiplex immunohistochemistry/immunofluorescence techniques in the era of cancer immunotherapy. Cancer Commun (Lond) 2020; 40:135-153. [PMID: 32301585 PMCID: PMC7170662 DOI: 10.1002/cac2.12023] [Citation(s) in RCA: 325] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/20/2020] [Indexed: 12/17/2022] Open
Abstract
Conventional immunohistochemistry (IHC) is a widely used diagnostic technique in tissue pathology. However, this technique is associated with a number of limitations, including high inter-observer variability and the capacity to label only one marker per tissue section. This review details various highly multiplexed techniques that have emerged to circumvent these constraints, allowing simultaneous detection of multiple markers on a single tissue section and the comprehensive study of cell composition, cellular functional and cell-cell interactions. Among these techniques, multiplex Immunohistochemistry/Immunofluorescence (mIHC/IF) has emerged to be particularly promising. mIHC/IF provides high-throughput multiplex staining and standardized quantitative analysis for highly reproducible, efficient and cost-effective tissue studies. This technique has immediate potential for translational research and clinical practice, particularly in the era of cancer immunotherapy.
Collapse
Affiliation(s)
- Wei Chang Colin Tan
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore169856Singapore
| | | | - Hai Yun Cai
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore169856Singapore
| | - Harry Ho Man Ng
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
- Duke‐NUS Medical SchoolSingapore169856Singapore
| | - Duoduo Wu
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore169856Singapore
| | - Yu Ting Felicia Wee
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR)Singapore169856Singapore
| | - Joe Yeong
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR)Singapore169856Singapore
- Singapore Immunology NetworkAgency of Science (SIgN)Technology and Research (A*STAR)Singapore169856Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
| |
Collapse
|
18
|
Yeong J, Tan T, Chow ZL, Cheng Q, Lee B, Seet A, Lim JX, Lim JCT, Ong CCH, Thike AA, Saraf S, Tan BYC, Poh YC, Yee S, Liu J, Lim E, Iqbal J, Dent R, Tan PH. Multiplex immunohistochemistry/immunofluorescence (mIHC/IF) for PD-L1 testing in triple-negative breast cancer: a translational assay compared with conventional IHC. J Clin Pathol 2020; 73:557-562. [PMID: 31969377 DOI: 10.1136/jclinpath-2019-206252] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) monoclonal antibody therapy has recently gained approval for treating metastatic triple-negative breast cancer (TNBC) -, in particular in the PD-L1+ patient subgroup of the recent IMpassion130 trial. The SP142 PD-L1 antibody clone was used as a predictive assay in this trial, but this clone was found to be an outlier in previous harmonisation studies in lung cancer. AIMS To address the comparability of PD-L1 clones in TNBC, we evaluated the concordance between conventional immunohistochemistry (IHC) and multiplex immunohistochemistry/immunofluorescence (mIHC/IF) that allowed simultaneous quantification of three different PD-L1 antibodies (22C3, SP142 and SP263). METHODS Our cohort comprised 25 TNBC cases, 12 non-small-cell lung carcinomas and 8 other cancers. EpCAM labelling was used to distinguish tumour cells from immune cells. RESULTS Moderate-to-strong correlations in PD-L1 positivity were found between results obtained through mIHC/IF and IHC. Individual concordance rates in the study ranged from 67% to 100%, with Spearman's rank correlation coefficient values up to 0.88. CONCLUSIONS mIHC/IF represents a promising tool in the era of cancer immunotherapy, as it can simultaneously detect and quantify PD-L1 labelling with multiple antibody clones, and allow accurate evaluation of tumour and immune cells. Clinicians and pathologists require this information to predict patient response to anti-PD-1/PD-L1 therapy. The adoption of this assay may represent a significant advance in the management of therapeutically challenging cancers. Further analysis and assay harmonisation are essential for translation to a routine diagnostic setting.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore .,Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Tira Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Zi Long Chow
- Division of Pathology, Singapore General Hospital, Singapore.,University of Tasmania, Hobart, Tasmania, Australia
| | - Qing Cheng
- Duke-NUS Medical School, Duke-NUS Medical School, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Amanda Seet
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | | | - Jeffrey Chun Tatt Lim
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Clara Chong Hui Ong
- Division of Pathology, Singapore General Hospital, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, Singapore
| | - Sahil Saraf
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Yong Cheng Poh
- Diagnostics Development (DxD) Hub, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Sidney Yee
- Diagnostics Development (DxD) Hub, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Jin Liu
- Duke-NUS Medical School, Duke-NUS Medical School, Singapore
| | - Elaine Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, Singapore
| | - Rebecca Dent
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| |
Collapse
|
19
|
Lam JH, Ng HHM, Lim CJ, Sim XN, Malavasi F, Li H, Loh JJH, Sabai K, Kim JK, Ong CCH, Loh T, Leow WQ, Choo SP, Toh HC, Lee SY, Chan CY, Chew V, Lim TS, Yeong J, Lim TKH. Expression of CD38 on Macrophages Predicts Improved Prognosis in Hepatocellular Carcinoma. Front Immunol 2019; 10:2093. [PMID: 31552039 PMCID: PMC6738266 DOI: 10.3389/fimmu.2019.02093] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 08/20/2019] [Indexed: 12/28/2022] Open
Abstract
Background: CD38 is involved in the adenosine pathway, which represents one of the immunosuppressive mechanisms in cancer. CD38 is broadly expressed across immune cell subsets, including human macrophages differentiated in vitro from monocytes, but expression by tissue-resident macrophages remains to be demonstrated. Methods: Tissue samples were obtained from 66 patients with hepatocellular carcinoma (HCC) from Singapore and analyzed using immunohistochemistry. Tumor-infiltrating leukocytes (TILs) were further examined using DEPArray™, and the phenotype of freshly isolated TILs was determined using flow cytometry. Results: CD38 was frequently co-expressed with the macrophage-specific marker CD68. CD38+CD68+ macrophage density was associated with improved prognosis after surgery, while total CD68+ macrophage density was associated with poor prognosis. DEPArray™ analysis revealed the presence of large (>10 μm), irregularly shaped CD45+CD14+ cells that resembled macrophages, with concurrent CD38+ expression. Flow cytometry also revealed that majority of CD14+HLA-DR+ cells expressed CD38. Conclusion: CD38 expression was clearly demonstrated on human macrophages in an in vivo setting. The positive association identified between CD38+ macrophage density and prognosis may have implications for routine diagnostic work.
Collapse
Affiliation(s)
- Jian Hang Lam
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,A. Menarini Biomarkers Singapore Pte Ltd., Singapore, Singapore
| | | | - Chun Jye Lim
- Translational Immunology Institute (TII), SingHealth DukeNUS Academic Medical Centre, Singapore, Singapore
| | - Xin Ni Sim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,Temasek Polytechnic, Singapore, Singapore
| | - Fabio Malavasi
- Department of Medical Science, University of Turin and Fondazione Ricerca Molinette, Turin, Italy
| | - Huihua Li
- Division of Medicine, Singapore General Hospital, Singapore, Singapore.,Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Josh Jie Hua Loh
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Khin Sabai
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joo-Kyung Kim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | | | - Tracy Loh
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Wei Qiang Leow
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Ser Yee Lee
- Duke-NUS Medical School, Singapore, Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Chung Yip Chan
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth DukeNUS Academic Medical Centre, Singapore, Singapore
| | - Tong Seng Lim
- A. Menarini Biomarkers Singapore Pte Ltd., Singapore, Singapore
| | - Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,Institute of Molecular Cell Biology, Agency of Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Tony Kiat Hon Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
20
|
Evaluation of phospho-histone H3 in Asian triple-negative breast cancer using multiplex immunofluorescence. Breast Cancer Res Treat 2019; 178:295-305. [PMID: 31410680 DOI: 10.1007/s10549-019-05396-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/04/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE We used multiplex immunofluorescence (mIF) to determine whether mitotic rate represents an independent prognostic marker in triple-negative breast cancer (TNBC). Secondary aims were to confirm the prognostic significance of immune cells in TNBC, and to investigate the relationship between immune cells and proliferating tumour cells. METHODS A retrospective Asian cohort of 298 patients with TNBC diagnosed from 2003 to 2015 at the Singapore General Hospital was used in the present study. Formalin-fixed, paraffin-embedded breast cancer samples were analysed on tissue microarrays using mIF, which combined phospho-histone H3 (pHH3) expression with cytokeratin (CK) and leukocyte common antigen (CD45) expression to identify tumour and immune cells, respectively. RESULTS Multivariate analysis showed that a high pHH3 index was associated with significantly improved overall survival (OS; p = 0.004), but this was not significantly associated with disease-free survival (DFS; p = 0.22). Similarly, multivariate analysis also revealed that a pHH3 positive count of > 1 cell per high-power field in the malignant epithelial compartment was an independent favourable prognostic marker for OS (p = 0.033) but not for DFS (p = 0.250). Furthermore, a high CD45 index was an independent favourable prognostic marker for DFS (p = 0.018), and there was a significant positive correlation between CD45 and pHH3 index (Spearman rank correlation coefficient, 0.250; p < 0.001). CONCLUSIONS Mitotic rates as determined by pHH3 expression in epithelial cells are significantly associated with improved survival in TNBC. mIF analysis of pHH3 in combination with CK and CD45 could help clinicians in prognosticating patients with TNBC.
Collapse
|
21
|
The role of Ki-67 in Asian triple negative breast cancers: a novel combinatory panel approach. Virchows Arch 2019; 475:709-725. [PMID: 31407032 DOI: 10.1007/s00428-019-02635-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/14/2019] [Accepted: 07/24/2019] [Indexed: 12/23/2022]
Abstract
The proliferation marker Ki-67 is frequently used to assess aggressiveness in the pathological evaluation of cancer, but its role remains uncertain in triple-negative breast cancer (TNBC). We aimed to quantify and localize Ki-67 expression in both epithelial and immune compartments in TNBC and investigate its association with clinicopathological parameters and survival outcomes. A total of 406 TNBC cases diagnosed between 2003 and 2015 at Singapore General Hospital were recruited. Using state-of-the-art, 7-colour multiplex immunofluorescence (mIF) tissue microarrays (TMAs) were stained to assess the abundance, density and spatial distribution of Ki-67-positive tumour cells and immune cells co-decorated with cytokeratin (CK) and leukocyte common antigen (CD45) respectively. Furthermore, MKI67 mRNA profiles were analysed using NanoString technology. In multivariate analysis adjusted for tumour size, histologic grade, age at diagnosis, and lymph node stage, a high Ki-67 labelling index (LI) > 0.3% was associated with improved disease-free survival (DFS; HR = 0.727; p = 0.027). High Ki-67-positive immune cell count per TMA was a favourable prognostic marker for both DFS (HR = 0.379; p = 0.00153) and overall survival (OS; HR = 0.473; p = 0.0482). The combination of high Ki-67 LI and high MKI67 expression was associated with improved DFS (HR = 0.239; p = 0.00639) and OS (HR = 0.213; p = 0.034). This study is among the first to highlight that Ki-67 is associated with favourable prognosis in an adjuvant setting in TNBC, and the mIF-based evaluation of Ki-67 expression on both tumour and immune cells represents a novel prognostic approach.
Collapse
|
22
|
Scrima AT, Lubner MG, Abel EJ, Havighurst TC, Shapiro DD, Huang W, Pickhardt PJ. Texture analysis of small renal cell carcinomas at MDCT for predicting relevant histologic and protein biomarkers. Abdom Radiol (NY) 2019; 44:1999-2008. [PMID: 29804215 DOI: 10.1007/s00261-018-1649-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To assess CT texture features of small renal cell carcinomas (≤ 4cm) for association with key pathologic features including protein biomarkers. METHODS Quantitative CT texture analysis (CTTA) of small renal cancers (≤ 4cm) was performed on non-contrast and portal venous phase abdominal MDCT scans with an ROI drawn at the largest cross-sectional diameter of the tumor using commercially available software. Texture parameters including mean pixel attenuation, the standard deviation (SD) of the pixel distribution histogram, entropy, the mean of positive pixels, the skewness (i.e., asymmetry) of the pixel histogram, kurtosis (i.e., peakness) of the pixel histogram, and the percentage of positive pixels were correlated with pathologic data from surgical resection, including histology and nuclear grade, as well as microarray analysis in a subset (n = 40) including Ki67 index, CRP, and neovascularization (CD105/CD31). RESULTS Portal venous phase images were available in 249 patients (105 women, 144 men; mean age, 56.7 years) with tumors ≤ 4cm (mean, median, range, ± SD; 2.66, 2.60, 0.3-4.0 ± 0.85 cm). CT texture features of standard deviation, mean of the positive pixels, and entropy of the pixel histogram were significantly associated with histologic cell type (clear vs. non-clear; p < 0.001). Entropy and mean of the positive pixels also showed an association with nuclear grade, although not statistically significant. In the microarray analysis subset, kurtosis of the pixel histogram was associated with CD105/CD31 (p = 0.05). SD also showed some association with CD 105 positivity (p = 0.02) and CAIX expression (p = 0.01). Non-contrast CT images were available in 174 patients (72 women, 102 men; mean age, 57.5 years). Although the association with histology was not as strong as on the portal venous phase, in the subset of patients with microarray data, SD was found to correlate with CRP (p = 0.08), kurtosis with CRP (p = 0.004), CD105/CD31 (p = 0.002), and with Ki 67 index (p < 0.001). CONCLUSION CT texture features were significantly associated with important histopathologic features in small renal cancers. These non-invasive measures can be performed retrospectively and may provide useful information when determining follow-up and treatment of small renal cancers.
Collapse
Affiliation(s)
- Andrew T Scrima
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Meghan G Lubner
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA.
| | - E Jason Abel
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Thomas C Havighurst
- Department of Biostatistics, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Daniel D Shapiro
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Wei Huang
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| |
Collapse
|
23
|
Yeong J, Lim JCT, Lee B, Li H, Ong CCH, Thike AA, Yeap WH, Yang Y, Lim AYH, Tay TKY, Liu J, Wong SC, Chen J, Lim EH, Iqbal J, Dent R, Newell EW, Tan PH. Prognostic value of CD8 + PD-1+ immune infiltrates and PDCD1 gene expression in triple negative breast cancer. J Immunother Cancer 2019; 7:34. [PMID: 30728081 PMCID: PMC6366051 DOI: 10.1186/s40425-019-0499-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
The role of programmed cell death protein-1 (PD-1)/programmed cell death ligand 1 (PD-L1) in triple negative breast cancer (TNBC) remains to be fully understood. In this study, we investigated the role of PD-1 as a prognostic marker for TNBC in an Asian cohort (n = 269). Samples from patients with TNBC were labeled with antibodies against PD-L1 and PD-1, and subjected to NanoString assays to measure the expression of immune-related genes. Associations between disease-free survival (DFS), overall survival (OS) and biomarker expression were investigated. Multivariate analysis showed that tumors with high PD-1+ immune infiltrates harbored significantly increased DFS, and this increase was significant even after controlling for clinicopathological parameters (HR 0.95; P = 0.030). In addition, the density of cells expressing both CD8 and PD-1, but not the density of CD8−PD-1+ immune infiltrates, was associated with improved DFS. Notably, this prognostic significance was independent of clinicopathological parameters and the densities of total CD8+ cell (HR 0.43, P = 0.011). At the transcriptional level, high expression of PDCD1 within the tumor was significantly associated with improved DFS (HR 0.38; P = 0.027). In line with these findings, high expression of IFNG (HR 0.38; P = 0.001) and IFN signaling genes (HR 0.46; p = 0.027) was also associated with favorable DFS. Inclusion of PD-1 immune infiltrates and PDCD1 gene expression added significant prognostic value for DFS (ΔLRχ2 = 6.35; P = 0.041) and OS (ΔLRχ2 = 9.53; P = 0.008), beyond that provided by classical clinicopathological variables. Thus, PD-1 mRNA and protein expression status represent a promising, independent indicator of prognosis in TNBC.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Jeffrey Chun Tatt Lim
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Huihua Li
- Division of Medicine, Singapore General Hospital, Singapore, Singapore
| | - Clara Chong Hui Ong
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Wei Hseun Yeap
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Yi Yang
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore.,Shanghai University of Finance and Economics, Shanghai, China
| | - Ansel Yi Herh Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Timothy Kwang Yong Tay
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore
| | - Jin Liu
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Siew-Cheng Wong
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore
| | - Elaine Hsuen Lim
- National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Rebecca Dent
- National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore.
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), 8A, Biomedical Grove, Immunos, Singapore, 138648, Singapore.
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, 20 College Road, Academia, Level 7, Singapore, 169856, Singapore. .,Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
24
|
Wickenhauser C, Bethmann D, Feng Z, Jensen SM, Ballesteros-Merino C, Massa C, Steven A, Bauer M, Kaatzsch P, Pazaitis N, Toma G, Bifulco CB, Fox BA, Seliger B. Multispectral Fluorescence Imaging Allows for Distinctive Topographic Assessment and Subclassification of Tumor-Infiltrating and Surrounding Immune Cells. Methods Mol Biol 2019; 1913:13-31. [PMID: 30666596 DOI: 10.1007/978-1-4939-8979-9_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Histomorphology has significantly changed over the last decades due to technological achievements in immunohistochemistry (IHC) for the visualization of specific proteins and in molecular pathology, particularly in the field of in situ hybridization of small oligonucleotides and amplification of DNA and RNA amplicons. With an increased availability of suitable methods, the demands regarding the observer of histomorphological slides were the supply of complex quantitative data as well as more information about protein expression and cell-cell interactions in tissue sections. Advances in fluorescence-based multiplexed IHC techniques, such as multispectral imaging (MSI), allow the quantification of multiple proteins at the same tissue section. In histopathology, it is a well-known technique for over a decade yet harboring serious problems concerning quantitative preciseness and tissue autofluorescence of multicolor staining when using formalin-fixed, paraffin-embedded (FFPE) tissue specimen. In recent years, milestones in tissue preparation, fluorescent dyes, hardware imaging, and software analysis were achieved including automated tissue segmentation (e.g., tumor vs. stroma) as well as in cellular and subcellular multiparameter analysis.This chapter covers the role that MSI plays in anatomic pathology for the analysis of FFPE tissue sections, discusses the technical aspects of MSI, and provides a review of its application in the characterization of immune cell infiltrates and beyond regarding its prognostic and predictive value and its use for guidance of clinical decisions for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Claudia Wickenhauser
- Medical Faculty, Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Daniel Bethmann
- Medical Faculty, Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Zipei Feng
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Shawn M Jensen
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Carmen Ballesteros-Merino
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Chiara Massa
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andre Steven
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Marcus Bauer
- Medical Faculty, Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Kaatzsch
- Medical Faculty, Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Nikolaos Pazaitis
- Medical Faculty, Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Georgiana Toma
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Carlo B Bifulco
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Bernard A Fox
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
25
|
An integrated automated multispectral imaging technique that simultaneously detects and quantitates viral RNA and immune cell protein markers in fixed sections from Epstein-Barr virus-related tumours. Ann Diagn Pathol 2018; 37:12-19. [PMID: 30218928 DOI: 10.1016/j.anndiagpath.2018.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/03/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM Epstein-Barr virus (EBV) is an oncovirus that is commonly associated with the development of lymphomas and epithelial carcinomas. In the era of immunotherapy, histological evaluation of EBV-related cancers is currently a multi-sample, multi-technique process requiring separate time-consuming detection of EBV-encoded small RNAs by in situ hybridisation (ISH), and parallel labelling of sections for cancer-associated protein markers. METHODS Using EBV-associated tumours as proof-of-concept for feasibility, here we developed an approach that allows simultaneous detection of EBV RNAs and multiple protein markers such as PD-L1, EBV-LMP, CD8, CD4, CD20, CD30 and CD15on a single tissue section based on our recently reported automated staining protocol. RESULTS We successfully combined multiplex immunofluorescence (mIF) to detect 3 abovementioned protein markers involved in cancer, with ISH, and applied the protocol to f tissue samples from patients diagnosed with EBV-associated pulmonary lymphoepithelioma-like carcinoma (LELC), gastric carcinoma and Hodgkin's Lymphoma. Empowered by the Vectra 3 Automated Quantitative Pathology Imaging System, we demonstrate the utility and potential of this integrated approach to concurrently detect and quantitate viral RNA and protein biomarkers of immune and tumour cells. CONCLUSION This study represents an important step forward in the research and diagnosis of EBV-associated cancers, and could be readily modified to include other proteins and RNA markers to apply to other malignancies. More importantly, the novel automated ISH-mIF protocol that we detailly described here could also be readily reproduced by most of the diagnostic and research lab to future projects that aim to look at both RNA and protein markers.
Collapse
|
26
|
Moreno Roig E, Yaromina A, Houben R, Groot AJ, Dubois L, Vooijs M. Prognostic Role of Hypoxia-Inducible Factor-2α Tumor Cell Expression in Cancer Patients: A Meta-Analysis. Front Oncol 2018; 8:224. [PMID: 29942795 PMCID: PMC6004384 DOI: 10.3389/fonc.2018.00224] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-inducible factor-2α (HIF-2α) plays an important role in tumor progression and metastasis. A number of studies have evaluated the correlation between HIF-2α overexpression and clinical outcome in cancer patients but yielded inconsistent results. To comprehensively and quantitatively summarize the evidence on the capability of HIF-2α to predict the prognosis of cancer patients with solid tumors, a meta-analysis was carried out. Renal cell carcinoma (CC-RCC) was separately analyzed due to an alternative mechanism of regulation. Systematic literature searches were performed in PubMed and Embase databases for relevant original articles until February 2018. Forty-nine studies with 6,052 patients were included in this study. The pooled hazard ratios (HRs) with corresponding confidence intervals were calculated to assess the prognostic value of HIF-2α protein expression in tumor cells. The meta-analysis revealed strong significant negative associations between HIF-2α expression and five endpoints: overall survival [HR = 1.69, 95% confidence interval (95% CI) 1.39-2.06], disease-free survival (HR = 1.87, 95% CI 1.2-2.92), disease-specific survival (HR = 1.57, 95% CI 1.06-2.34), metastasis-free survival (HR = 2.67, 95% CI 1.32-5.38), and progression-free survival (HR = 2.18, 95% CI 1.25-3.78). Subgroup analyses revealed similar associations in the majority of tumor sites. Overall, these data demonstrate a negative prognostic role of HIF-2α in patients suffering from different types of solid tumors.
Collapse
Affiliation(s)
- Eloy Moreno Roig
- Department of Radiotherapy (MAASTRO)/GROW - School for Developmental Biology and Oncology, Maastricht University, Maastricht, Netherlands
| | - Ala Yaromina
- Department of Radiotherapy (MAASTRO)/GROW - School for Developmental Biology and Oncology, Maastricht University, Maastricht, Netherlands
| | - Ruud Houben
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, Netherlands
| | - Arjan J Groot
- Department of Radiotherapy (MAASTRO)/GROW - School for Developmental Biology and Oncology, Maastricht University, Maastricht, Netherlands
| | - Ludwig Dubois
- Department of Radiotherapy (MAASTRO)/GROW - School for Developmental Biology and Oncology, Maastricht University, Maastricht, Netherlands
| | - Marc Vooijs
- Department of Radiotherapy (MAASTRO)/GROW - School for Developmental Biology and Oncology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVEIW Approximately one in three patients with nonmetastatic renal cell carcinoma (RCC) at the time of surgery will subsequently develop local or metastatic recurrence. The purpose of this review is to examine the current rationale for surveillance, describe sites of RCC metastasis, evaluate the existing guidelines for postsurgical follow-up studies, and analyze the risk stratification systems following RCC surgery. RECENT FINDINGS Although 75% of recurrences will be identified during the first 5 years following surgery, late recurrences are not uncommon. The risk of recurrence can be predicted from the tumor stage, grade, and other pathologic features. Advanced risk stratification will likely be possible in the future with increased use of molecular classification and serum biomarkers. Patient comorbidities, age, and individual recurrence risk should also be considered when designing individualized surveillance protocols. SUMMARY Follow-up after surgery for RCC should focus on imaging of the chest and abdomen to detect common sites of recurrence. Patients should be stratified for risk, and surveillance imaging should be more frequent and intensive in healthy patients with higher risk. Future research is needed to define an optimal individualized surveillance strategy that balances the potential benefits of early cancer detection with the risks and cost of surveillance.
Collapse
|
28
|
Lim JCT, Yeong JPS, Lim CJ, Ong CCH, Wong SC, Chew VSP, Ahmed SS, Tan PH, Iqbal J. An automated staining protocol for seven-colour immunofluorescence of human tissue sections for diagnostic and prognostic use. Pathology 2018; 50:333-341. [PMID: 29429740 DOI: 10.1016/j.pathol.2017.11.087] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023]
Abstract
Multiplex immunofluorescence (mIF) allows simultaneous antibody-based detection and quantification of the expression of up to six markers, plus a nuclear counterstain, on a single tissue section. Recent studies have shown the potential for mIF to advance our understanding of complex disease processes, including cancer. It is important that the technique be standardised and validated to facilitate its transition into clinical use. Traditional approaches to mIF rely on manual processing of sections, which is time-consuming and a source of significant variation between samples/individuals. Here we determined if an automated diagnostic tissue stainer could be used for mIF incorporating tyramide signal amplification (TSA), and how the final image quality compared with sections stained semi-automatically or manually. Using tissue microarrays of fixed human breast tumour sections, we observed comparable antibody labelling between the diagnostic autostainer and manual technique. The diagnostic autostainer produced higher signal intensity with similar spectral unmixing efficiency. We also found that microwave treatment for antibody stripping during TSA labelling could be replaced by the heating option incorporated within the diagnostic-use autostainer. These data show that diagnostic autostainers used for traditional immunohistochemistry protocols can be readily adapted to achieve rapid preparation of high-quality sections using a TSA method for clinical mIF.
Collapse
Affiliation(s)
| | - Joe Poh Sheng Yeong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore; Singapore Immunology Network, Agency of Science, Technology and Research, Singapore
| | - Chun Jye Lim
- SingHealth Translational Immunology and Inflammation Centre, Singapore
| | | | - Siew Cheng Wong
- Singapore Immunology Network, Agency of Science, Technology and Research, Singapore
| | | | | | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore
| | - Jabed Iqbal
- Department of Anatomical Pathology, Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore.
| |
Collapse
|
29
|
Li J, Liao Y, Ding T, Wang B, Yu X, Chu Y, Xu J, Zheng L. Tumor-infiltrating macrophages express interleukin-25 and predict a favorable prognosis in patients with gastric cancer after radical resection. Oncotarget 2017; 7:11083-93. [PMID: 26840565 PMCID: PMC4905459 DOI: 10.18632/oncotarget.7095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 01/17/2016] [Indexed: 12/28/2022] Open
Abstract
Interleukin-25 (IL-25) is a recently identified member of the proinflammatory IL-17 cytokine family; however, its role in human tumors remains largely unknown. The aim of this study was to investigate the cellular source and clinical significance of IL-25 in gastric cancer (GC) in situ. The results demonstrated that macrophages (Mφs) were the primary IL-25-expressing cells (IL-25+) in GC in situ. Moreover, IL-25+ cells were highly enriched in the intra-tumoral (IT) region of GC tissues (p < 0.001). The production of IL-25 in Mφs exposed to culture supernatant from gastric cancer cell line SGC7901 in vitro was induced by transforming growth factor-β1, and their density in the IT region was positively associated with those of other effector immune cells, namely, CD4+ T cells, CD8+ T cells and CD103+T cells (p < 0.01). This suggested that macrophages might produce IL-25 to create an antitumor micromilieu in GC tissues. The level of IL-25+IT cells was positively associated with histological grade (p < 0.001) and found to be an independent predictor of favorable survival (p = 0.024) in patients with GC after radical resection. These findings suggest that IL-25+IT cells may be a novel therapeutic target in those patients.
Collapse
Affiliation(s)
- Jinqing Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yuan Liao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.,Department of Laboratory Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Tong Ding
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Bo Wang
- Department of Urology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xingjuan Yu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yifan Chu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jing Xu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Limin Zheng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.,Key Laboratory of Gene Engineering of The Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
30
|
Bellut J, Bertz S, Nolte E, Stöhr C, Polifka I, Lieb V, Herrmann E, Jung R, Hartmann A, Wullich B, Taubert H, Wach S. Differential prognostic value of MYC immunohistochemistry in subtypes of papillary renal cell carcinoma. Sci Rep 2017; 7:16424. [PMID: 29180625 PMCID: PMC5703709 DOI: 10.1038/s41598-017-16144-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/08/2017] [Indexed: 01/21/2023] Open
Abstract
The histomorphological subtyping of papillary renal cell carcinomas (pRCCs) has improved the predictions of patients' long-term survival. Based on our previous results, we hypothesized that the MYC proto-oncogene would show differential expression in pRCC subtypes. Using a multi-institutional cohort of 204 pRCC patients we assessed the additional value of the immunohistochemical markers MYC, MINA53, and Ki67 in predicting patient's long-term survival. The clinical endpoints were overall survival (OS) and cancer-specific survival (CSS). Nomograms were constructed to predict each patient's risk of death (OS). The incorporation of the MYC staining patterns allowed the stratification of pRCC type 1 patients into better and worse prognostic groups. None of the patients with pRCC type 1 tumors and favorable MYC staining patterns died from tumor-related causes. This prognostic value was independent of the patient's age at surgery, the pathological tumor stage and presence of lymph node invasion. we could show that the immunohistochemical assessment of MYC and the histomorphological subtyping of pRCC stratifies pRCC type 1 tumors with regard to OS and CSS. The determination of the histomorphologic pRCC subtype in combination with the MYC immunohistochemical staining patterns allows a more accurate prediction of patients' individual risk of death.
Collapse
Affiliation(s)
- Julia Bellut
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Bertz
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Elke Nolte
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Christine Stöhr
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Iris Polifka
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Verena Lieb
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | | | - Rudolf Jung
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Bernd Wullich
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Helge Taubert
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany.
| | - Sven Wach
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
31
|
Marcelin C, Ambrosetti D, Bernhard J, Roy C, Grenier N, Cornelis F. Percutaneous image-guided biopsies of small renal tumors: Current practice and perspectives. Diagn Interv Imaging 2017; 98:589-599. [DOI: 10.1016/j.diii.2017.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 07/19/2017] [Accepted: 07/24/2017] [Indexed: 12/30/2022]
|
32
|
Donovan MJ, Cordon-Cardo C. Implementation of a Precision Pathology Program Focused on Oncology-Based Prognostic and Predictive Outcomes. Mol Diagn Ther 2017; 21:115-123. [PMID: 28000172 DOI: 10.1007/s40291-016-0249-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Personalized or precision medicine as a diagnostic and therapeutic paradigm was introduced some 10-15 years ago, with the advent of biomarker discovery as a mechanism for identifying prognostic and predictive attributes associated with treatment indication and outcome. While the concept is not new, the successful development and implementation of novel 'companion diagnostics', especially in oncology, continues to represent a significant challenge and is currently at the forefront of smart trial design and therapeutic choice. The ability to determine patient selection for a specific therapy has broad implications including better chances for a positive outcome, limited exposure to potentially toxic drugs and improved health economics. Importantly, a significant step in this paradigm is the role of predictive pathology or the accurate assessment of morphology at the microscopic level. In breast cancer, this has been most useful where histologic attributes such as the classification of tubular and cribriform carcinoma dictates surgery while neoadjuvant studies suggest that patients with lobular carcinoma are not likely to benefit from chemotherapy. The next level of 'personalized pathology' at the tissue-cellular level is the use of 'protein biomarker panels' to classify the disease process and ultimately drive tumor characterization and treatment. The following review article will focus on the evolution of predictive pathology from a subjective, 'opinion-based' approach to a quantitative science. In addition, we will discuss the individual components of the precise pathology platform including advanced image analysis, biomarker quantitation with mathematical modeling and the integration with fluid-based (i.e. blood, urine) analytics as drivers of next generation precise patient phenotyping.
Collapse
|
33
|
Yeong J, Thike AA, Lim JCT, Lee B, Li H, Wong SC, Hue SSS, Tan PH, Iqbal J. Higher densities of Foxp3+ regulatory T cells are associated with better prognosis in triple-negative breast cancer. Breast Cancer Res Treat 2017; 163:21-35. [DOI: 10.1007/s10549-017-4161-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/13/2017] [Indexed: 01/29/2023]
|
34
|
Garnelo M, Tan A, Her Z, Yeong J, Lim CJ, Chen J, Lim KH, Weber A, Chow P, Chung A, Ooi LLPJ, Toh HC, Heikenwalder M, Ng IOL, Nardin A, Chen Q, Abastado JP, Chew V. Interaction between tumour-infiltrating B cells and T cells controls the progression of hepatocellular carcinoma. Gut 2017; 66:342-351. [PMID: 26669617 PMCID: PMC5284473 DOI: 10.1136/gutjnl-2015-310814] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The nature of the tumour-infiltrating leucocytes (TILs) is known to impact clinical outcome in carcinomas, including hepatocellular carcinoma (HCC). However, the role of tumour-infiltrating B cells (TIBs) remains controversial. Here, we investigate the impact of TIBs and their interaction with T cells on HCC patient prognosis. DESIGN Tissue samples were obtained from 112 patients with HCC from Singapore, Hong Kong and Zurich and analysed using immunohistochemistry and immunofluorescence. RNA expression of CD19, CD8A, IFNG was analysed using quantitative PCR. The phenotype of freshly isolated TILs was analysed using flow cytometry. A mouse model depleted of mature B cells was used for functional study. RESULTS Tumour-infiltrating T cells and B cells were observed in close contact with each other and their densities are correlated with superior survival in patients with HCC. Furthermore, the density of TIBs was correlated with an enhanced expression of granzyme B and IFN-γ, as well as with reduced tumour viability defined by low expression of Ki-67, and an enhanced expression of activated caspase-3 on tumour cells. CD27 and CD40 costimulatory molecules and TILs expressing activation marker CD38 in the tumour were also correlated with patient survival. Mice depleted of mature B cells and transplanted with murine hepatoma cells showed reduced tumour control and decreased local T cell activation, further indicating the important role of B cells. CONCLUSIONS The close proximity of tumour-infiltrating T cells and B cells indicates a functional interaction between them that is linked to an enhanced local immune activation and contributes to better prognosis for patients with HCC.
Collapse
Affiliation(s)
- Marta Garnelo
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Alex Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Biopolis, Singapore
| | - Joe Yeong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore,Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Chun Jye Lim
- SingHealth Translational Immunology and Inflammation Centre (STIIC), Singapore Health Services Pte Ltd, Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Achim Weber
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Pierce Chow
- National Cancer Centre, Singapore, Singapore,Singapore General Hospital, Singapore, Singapore,Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Alexander Chung
- National Cancer Centre, Singapore, Singapore,Singapore General Hospital, Singapore, Singapore
| | - London Lucien PJ Ooi
- National Cancer Centre, Singapore, Singapore,Singapore General Hospital, Singapore, Singapore,Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany,Institute of Virology, Technical University München/Helmholtz Zentrum München, Germany
| | - Irene O L Ng
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Alessandra Nardin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Biopolis, Singapore,National Cancer Centre, Singapore, Singapore
| | - Jean-Pierre Abastado
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore,Institut de Recherches Internationales Servier, Suresnes, France
| | - Valerie Chew
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore,SingHealth Translational Immunology and Inflammation Centre (STIIC), Singapore Health Services Pte Ltd, Singapore, Singapore,Duke-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
35
|
Neovascularity as a prognostic marker in renal cell carcinoma. Hum Pathol 2016; 57:98-105. [PMID: 27436827 DOI: 10.1016/j.humpath.2016.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/20/2016] [Accepted: 07/02/2016] [Indexed: 12/13/2022]
Abstract
Endothelial markers platelet and endothelial cell adhesion molecule (PECAM-1), cluster of differentiation (CD31) and endoglin (CD105) may be used to identify endothelium and activated endothelium, respectively, with the CD105/CD31 ratio used to measure neovascularity. This study investigated the hypothesis that neovascularity in renal cell carcinoma (RCC) is associated with more aggressive RCC tumors and can be used to predict oncological outcomes. Multiplexed immunohistochemistry using antibodies to detect endoglin and PECAM-1 was performed on tissue microarray of benign kidney samples and RCC tumors including clear cell, papillary, chromophobe, and collecting duct and unclassified tumors (combined for statistics), and multispectral imaging was used for analysis. The CD105/CD31 ratio was compared with clinical and pathologic features of RCC as well as clinical outcomes after surgery using Cox proportional hazards regression and Kaplan-Meier analysis. A total of 502 tumor samples and 122 normal kidney samples from 251 RCC patients were analyzed. The average CD105/CD31 expression ratio, an indicator of neovascularization, was increased in higher pathologic stage tumors (P< .0001). Among RCC morphotypes, the ratio was lower in papillary RCC morphotype tumors (P= .001) and higher in collecting duct/unclassified tumors (P= .0001) compared with clear cell RCC. Among nuclear grades, grade 4 RCC displayed a significantly elevated CD105/CD31 ratio (P< .0001). In multivariable analysis, increased neovascularity was associated with decreased overall survival (hazard ratio, 1.54 [95% confidence interval, 1.06-2.23]; P= .02). In patients receiving anti-vascular endothelial growth factor therapy (VEGF, n = 13) for metastatic RCC, a low CD105/CD31 ratio was associated with increased survival (P= .02). We conclude that higher neovascularity is associated with worse outcomes after surgery for RCC. The ratio of CD105/CD31 expression is a potential indicator of response to anti-VEGF therapy.
Collapse
|
36
|
Abstract
Contemporary approaches to cancer therapy have moved away from a one-size-fits-all model. Precision treatments are based on patients and their individual tumor characteristics. In general, incidental renal masses fall into three categories: aggressive cancers, indolent cancers, and benign tumors. Treatments may include surgery, thermal ablation, or observation. Choosing the best treatment based solely on radiologic information is uninformed and unnecessary, and may lead to overtreatment of benign tumors or inappropriate treatment of aggressive tumors. Percutaneous biopsy is a safe and effective tool that provides prognostic information about unknown masses to guide treatment decision making. The first step for improving personalized treatments for small renal masses is clear: increase utilization of pretreatment renal mass biopsy.
Collapse
|
37
|
Krabbe LM, Margulis V, Lotan Y. Prognostic Role of Cell Cycle and Proliferative Markers in Clear Cell Renal Cell Carcinoma. Urol Clin North Am 2016; 43:105-18. [DOI: 10.1016/j.ucl.2015.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
38
|
Combined Angiogenesis and Proliferation Markers' Expressions as Long-Term Prognostic Factors in Renal Cell Cancer. Clin Genitourin Cancer 2015; 14:e283-9. [PMID: 26821530 DOI: 10.1016/j.clgc.2015.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the expression of MIB-1, BCL-2, VEGFR3, and CD31 and their associations with long-term survival in patients with renal cell cancer (RCC). PATIENTS AND METHODS This study consisted of 224 RCC patients who underwent radical nephrectomy from 1985 to 1995. Follow-up continued for up to over 20 years. MIB-1 and BCL-2 expression were analyzed alone, and additionally, the expression of MIB-1, BCL-2, VEGFR3, and CD31 were combined in pairs using the following groups: low/low, low/high, high/low, and high/high. RESULTS Low BCL-2 expression (hazard ratio [HR], 2.16; 95% confidence interval [CI], 1.42-3.31; P < .001 compared with high BCL-2 in univariate analysis) and high MIB-1 expression (HR, 2.05; 95% CI, 1.32-3.19; P = .001 in multivariate analysis) were found to associate for poorer survival in RCC. In multivariate analysis, the combination of high MIB-1/low BCL-2 was associated with poor survival compared with low MIB-1/high BCL-2 (HR, 3.20; 95% CI, 1.66-6.17; P = .001), and the combination of low VEGFR3/high CD31 was associated with poor survival (HR, 2.48; 95% CI, 1.29-4.78; P = .007) compared with high VEGFR3/high CD31. CONCLUSIONS Compared with high BCL-2 expression in combination with low or high MIB-1, VEGFR3, or CD31 expression, low BCL-2 expression in combination with low or high MIB-1, VEGFR3, or CD31 expression has poorer survival in the long-term follow-up of patients with RCC. Analysis of MIB-1, BCL-2, VEGFR3, and CD31 expression might be a useful additional marker to tailor the follow-up of RCC patients.
Collapse
|
39
|
Blute ML, Zorn K, Grimes M, Shi F, Downs TM, Jarrard DF, Best SL, Richards K, Nakada SY, Abel EJ. Extreme obesity does not predict poor cancer outcomes after surgery for renal cell cancer. BJU Int 2015; 118:399-407. [PMID: 26589741 DOI: 10.1111/bju.13381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To assess whether extreme obesity (body mass index [BMI] ≥ 40 kg/m(2) ) is associated with peri-operative outcomes, overall survival (OS), cancer-specific survival (CSS), or recurrence-free survival (RFS) after surgical treatment for renal cell carcinoma (RCC). PATIENTS AND METHODS After institutional review board approval, we used an institutional database to identify patients treated surgically between January 2000 and December 2014 with a pathological diagnosis of RCC. Comprehensive clinical and pathological data were reviewed. Kaplan-Meier analyses were used to estimate OS, RFS and CSS. Univariate and multivariate Cox proportional hazards analysis was used to evaluate associations with OS, CSS and RFS in patients with extreme obesity, among other known predictive variables. RESULTS In all, 100 patients (11.9%) with a BMI ≥ 40 kg/m(2) and 743 patients (88.1%) with a BMI < 40 kg/m(2) who were treated surgically for RCC were identified. Morbid obesity was not associated with an increased risk of blood transfusion (odds ratio [OR] 1, 95% confidence interval [CI] 0.587-1.70; P = 1.0). The median (interquartile range) length of hospital stay (LOS) was 4 (3-6) days. Morbid obesity was not associated with longer LOS (P = 0.26) or 30-day hospital readmission rates (P = 1.0). Major complications (Clavien ≥ 3a) were recorded in 67 patients (7.95%). BMI ≥ 40 kg/m(2) was not a predictor of major complications (OR 0.58, 95% CI 0.227-1.47; P = 0.251) or 90-day mortality (P = 0.4067). BMI ≥ 40 kg/m(2) was not associated with worse OS (P = 0.7), CSS (P = 0.2) or RFS (P = 0.5). BMI ≥ 35 kg/m(2) was also not associated with worse OS, CSS or RFS (P = 0.3, 0.1, 0.5, respectively). The 5-year OS rate was 68.9% for the entire cohort, including 69 and 70% for patients with BMI < 40 kg/m(2) and BMI ≥ 40 kg/m(2) , respectively (P = 0.69). The 5-year CSS was 79.5% for the entire cohort, including 78.4 and 87.9% (P = 0.16) for patients with BMI < 40 kg/m(2) and BMI ≥ 40 kg/m(2) , respectively. The 5-year RFS rates for BMI < 40 kg/m(2) and BMI ≥ 40 kg/m(2) were 84.1 and 90.6%, respectively (P = 0.48). CONCLUSIONS Extreme obesity is not associated with worse peri-operative or cancer outcomes after surgery for RCC. Surgery should remain a standard treatment option in well selected morbidly obese patients.
Collapse
Affiliation(s)
- Michael L Blute
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kristin Zorn
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Matthew Grimes
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Fangfang Shi
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tracy M Downs
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sara L Best
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kyle Richards
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Stephen Y Nakada
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - E Jason Abel
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
40
|
Bauman TM, Becka AJ, Sehgal PD, Huang W, Ricke WA. SIGIRR/TIR8, an important regulator of TLR4 and IL-1R-mediated NF-κB activation, predicts biochemical recurrence after prostatectomy in low-grade prostate carcinomas. Hum Pathol 2015; 46:1744-51. [PMID: 26344417 DOI: 10.1016/j.humpath.2015.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 01/08/2023]
Abstract
Single Ig IL-1-related receptor (SIGIRR) is a negative regulator of toll-like receptor 4 and IL-1-mediated activation of nuclear factor κ-light-chain enhancer of activated B cells. The purpose of this study was to qualitatively and quantitatively determine SIGIRR protein expression in human prostate tissues and associate SIGIRR expression with clinical parameters. SIGIRR expression was quantified in glandular prostate tissue using immunohistochemistry and multispectral imaging, and expression was evaluated in relation to clinicopathological features of benign prostatic hyperplasia and prostate cancer (PCa). Subgroupings of low Gleason score (≤ 6 and 3 + 4) and high Gleason score (4 + 3 and ≥ 8) were used for patient outcomes. SIGIRR was predominantly expressed in the cytoplasm and nucleus of the prostatic epithelium with little expression within the stroma. Compared with normal prostate, cytoplasmic SIGIRR expression was similar in benign prostatic hyperplasia, high-grade prostatic intraepithelial neoplasia, PCa, and metastases. A decrease in nuclear expression was found in metastasis samples (P = .04). Changes in SIGIRR expression were not associated with Gleason score, pathological stage, tumor volume, surgical margin status, or serum prostate-specific antigen (P > .05). Nuclear (P = .96) and cytoplasmic (P = .89) SIGIRR expressions were not related to patient outcomes in univariable analysis, but in the analysis of patients with low Gleason scores, high cytoplasmic SIGIRR expression was associated with biochemical recurrence in both univariable (P = .01) and multivariable (hazard ratio, 2.31 [95% confidence interval 1.05-5.06]; P = .04) analyses. Similarly, in multivariable analysis of only low-stage (pT2) tumors, SIGIRR independently predicted biochemical recurrence (P = .009). We conclude that SIGIRR predicts biochemical recurrence in patients with low Gleason score and low pathological stage PCa.
Collapse
Affiliation(s)
- Tyler M Bauman
- Division of Urologic Surgery, Department of Surgery, Washington University in St Louis School of Medicine, St Louis, MO
| | - Alexander J Becka
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Priyanka D Sehgal
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - William A Ricke
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI; George M. O'Brien Center, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
41
|
Bauman TM, Vezina CM, Huang W, Marker PC, Peterson RE, Ricke WA. Beta-catenin is elevated in human benign prostatic hyperplasia specimens compared to histologically normal prostate tissue. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:313-322. [PMID: 25606577 PMCID: PMC4297327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/09/2014] [Indexed: 06/04/2023]
Abstract
Benign prostatic hyperplasia (BPH) is linked to lower urinary tract symptoms (LUTS) such as incomplete bladder emptying, urinary frequency and urgency. Mechanisms responsible for BPH are not fully known. Here, we tested whether beta-catenin (CTNNB1) immunostaining intensity and distribution differ in human glandular BPH tissue specimens compared to normal prostate tissue. Multiplex immunostaining of CTNNB1, its putative transcriptional target gene lymphoid enhancer binding factor 1 (LEF1), and the epithelial marker E-cadherin were examined in clinical human prostate specimens with or without histological BPH (pure epithelial or mixed stromal-epithelial nodules). BPH specimens were obtained from 24 men who experienced LUTS and underwent transurethral resection of the prostate surgery. Control specimens were tumor-adjacent histologically normal prostate tissue from 48 patients who underwent radical prostatectomy. The resulting multispectral images were unmixed and optical densities recorded to quantify staining abundance, cellular (membranous, cytoplasmic, and nuclear) and tissue localization (stromal versus epithelial), and determination of percentage of CTNNB1-positive cells. The following CTNNB1 indices were significantly higher in BPH compared to normal prostate tissue: overall staining intensity, staining intensity in prostate stromal cell membranes, cytoplasm and nuclei, and prostate epithelial cell nuclei. The following LEF1 indices were significantly lower in BPH compared to tumor-adjacent normal prostate tissue: stromal LEF1 staining intensity, percentage of LEF1-positive stromal cells, and intensity of LEF1 staining in stromal cell membranes, cytoplasm, and nuclei. The percentage of stromal cells with CTNNB1(+)/LEF1(-) nuclei was higher and percentage of stromal cells with CTNNB1(-)/LEF1(+) nuclei was lower in BPH compared to tumor-adjacent normal prostate tissues. These results support the hypothesis that CTNNB1 expression increases in specific BPH tissue compartments. Further, since nuclear LEF1 staining does not coincide with cytoplasmic or nuclear CTNNB1 staining, it does not appear to be a reliable index of CTNNB1 activity in adult human prostate.
Collapse
Affiliation(s)
- Tyler M Bauman
- Department of Urology and Carbone Cancer Center, University of Wisconsin School of Medicine and Public HealthMadison, WI 53705, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary MedicineMadison, WI 53706, USA
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
| | - Wei Huang
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public HealthMadison, WI 53705, USA
| | - Paul C Marker
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
- Division of Pharmaceutical Sciences, University of Wisconsin School of PharmacyMadison, WI 53705, USA
| | - Richard E Peterson
- Division of Pharmaceutical Sciences, University of Wisconsin School of PharmacyMadison, WI 53705, USA
| | - William A Ricke
- Department of Urology and Carbone Cancer Center, University of Wisconsin School of Medicine and Public HealthMadison, WI 53705, USA
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
| |
Collapse
|
42
|
Donovan MJ, Cordon-Cardo C. Overcoming tumor heterogeneity in the molecular diagnosis of urological cancers. Expert Rev Mol Diagn 2014; 14:1023-31. [PMID: 25327491 DOI: 10.1586/14737159.2014.965151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Our understanding of tumor heterogeneity and impact on treatment response is still in its infancy, presenting significant challenges to the molecular pathologist, treating physician and ultimately for the patient. Given that tumor recurrence due to treatment resistance is the most common cause of cancer death, there remains a critical unmet need to change the current paradigm. The mechanisms which underlie tumor heterogeneity can be broadly divided into genomic instability and non-mutational processes, including stochastic variations in cellular responses, modulation by tumor microenvironment and or phenotypic/ functional plasticity relating to cancer stem cells. We believe that these biological mechanisms are not mutually exclusive and emphasize the need for more suitable methodologies to exploit the spatiotemporal patterns of intratumoral heterogeneity using novel approaches such as quantitative tissue-based biomarker assessment and systemic fluid analytics. Generating a comprehensive patient-centric phenotypic disease profile should generate a 'codex' which can be employed to change the current treatment decision process.
Collapse
Affiliation(s)
- Michael J Donovan
- Department of Pathology, Icahn School of Medicine, 1468 Madison Avenue, New York City, NY 10029, USA
| | | |
Collapse
|
43
|
Li L, Yan J, Xu J, Liu CQ, Zhen ZJ, Chen HW, Ji Y, Wu ZP, Hu JY, Zheng L, Lau WY. CXCL17 expression predicts poor prognosis and correlates with adverse immune infiltration in hepatocellular carcinoma. PLoS One 2014; 9:e110064. [PMID: 25303284 PMCID: PMC4193880 DOI: 10.1371/journal.pone.0110064] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/29/2014] [Indexed: 12/18/2022] Open
Abstract
CXC ligand 17 (CXCL17) is a novel CXC chemokine whose clinical significance remains largely unknown. In the present study, we characterized the prognostic value of CXCL17 in patients with hepatocellular carcinoma (HCC) and evaluated the association of CXCL17 with immune infiltration. We examined CXCL17 expression in 227 HCC tissue specimens by immunohistochemical staining, and correlated CXCL17 expression patterns with clinicopathological features, prognosis, and immune infiltrate density (CD4 T cells, CD8 T cells, B cells, natural killer cells, neutrophils, macrophages). Kaplan-Meier survival analysis showed that both increased intratumoral CXCL17 (P = 0.015 for overall survival [OS], P = 0.003 for recurrence-free survival [RFS]) and peritumoral CXCL17 (P = 0.002 for OS, P<0.001 for RFS) were associated with shorter OS and RFS. Patients in the CXCL17low group had significantly lower 5-year recurrence rate compared with patients in the CXCL17high group (peritumoral: 53.1% vs. 77.7%, P<0.001, intratumoral: 58.6% vs. 73.0%, P = 0.001, respectively). Multivariate Cox proportional hazards analysis identified peritumoral CXCL17 as an independent prognostic factor for both OS (hazard ratio [HR] = 2.066, 95% confidence interval [CI] = 1.296–3.292, P = 0.002) and RFS (HR = 1.844, 95% CI = 1.218–2.793, P = 0.004). Moreover, CXCL17 expression was associated with more CD68 and less CD4 cell infiltration (both P<0.05). The combination of CXCL17 density and immune infiltration could be used to further classify patients into subsets with different prognosis for RFS. Our results provide the first evidence that tumor-infiltrating CXCL17+ cell density is an independent prognostic factor that predicts both OS and RFS in HCC. CXCL17 production correlated with adverse immune infiltration and might be an important target for anti-HCC therapies.
Collapse
Affiliation(s)
- Li Li
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jing Yan
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jing Xu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chao-Qun Liu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Zuo-Jun Zhen
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China
| | - Huan-Wei Chen
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China
| | - Yong Ji
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China
| | - Zhi-Peng Wu
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China
| | - Jian-Yuan Hu
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China
| | - Limin Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wan Yee Lau
- Department of Hepatic and Pancreatic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, P. R. China; Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, P. R. China
| |
Collapse
|