1
|
Tanjak P, Chaiboonchoe A, Suwatthanarak T, Thanormjit K, Acharayothin O, Chanthercrob J, Parakonthun T, Methasate A, Fischer JM, Wong MH, Chinswangwatanakul V. Tumor-immune hybrid cells evade the immune response and potentiate colorectal cancer metastasis through CTLA4. Clin Exp Med 2024; 25:2. [PMID: 39499374 PMCID: PMC11538261 DOI: 10.1007/s10238-024-01515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 11/07/2024]
Abstract
Understanding the metastatic cascade is critical for the treatment and prevention of cancer-related death. Within a tumor, immune cells have the capacity to fuse with tumor cells to generate tumor-immune hybrid cells (THCs). THCs are hypothesized to be a subset of cancer cells with the capacity to enter circulation as circulating hybrid cells (CHC) and seed metastases. To understand the mechanism of THC metastasis, we investigated CHCs in peripheral blood from patients with stage IV colorectal cancer (CRC), as well as THCs in tissues of primary colorectal cancers and their liver metastasis sites using immunofluorescence, spatial proteomic, spatial transcriptomic, molecular classification, and molecular pathway analyses. Our findings indicated a high prevalence of CHCs and THCs in patients with stage IV CRC. THCs expressed CTLA4 in primary CRC lesions and correlated with upregulation of CD68, CD4, and HLA-DR in metastatic liver lesions, which is found in the consensus molecular subtype (CMS) 1 of primary CRC tissue. Pathway analysis of these genes suggested that THCs are associated with neutrophils due to upregulation of neutrophil extracellular trap signaling (NET) and neutrophil degranulation pathways. These data provide molecular pathways for the formation of THCs suggesting fusion with neutrophils, which may facilitate extravasation and metastatic seeding.
Collapse
Grants
- R016234003 Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
- R016234003 Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
- R016234003 Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
- R016234003 Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
- R016234003 Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- RO16241047 Foundation for Cancer Care, Siriraj Hospital, Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- 63-117 and 66-083 Health Systems Research Institute (HSRI), Thailand
- Mahidol University
Collapse
Affiliation(s)
- Pariyada Tanjak
- Faculty of Medicine Siriraj Hospital, Siriraj Cancer Center, Mahidol University, Bangkok, 10700, Thailand
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Amphun Chaiboonchoe
- Siriraj Center of Research Excellent for Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thanawat Suwatthanarak
- Faculty of Medicine Siriraj Hospital, Siriraj Cancer Center, Mahidol University, Bangkok, 10700, Thailand
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kullanist Thanormjit
- Faculty of Medicine Siriraj Hospital, Siriraj Cancer Center, Mahidol University, Bangkok, 10700, Thailand
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Onchira Acharayothin
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jantappapa Chanthercrob
- Siriraj Center of Research Excellent for Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thammawat Parakonthun
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Asada Methasate
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jared M Fischer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97201, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland , OR, 97201, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Melissa H Wong
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97201, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Vitoon Chinswangwatanakul
- Faculty of Medicine Siriraj Hospital, Siriraj Cancer Center, Mahidol University, Bangkok, 10700, Thailand.
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
2
|
Marcotte E, Goyeneche A, Abdouh M, Burnier JV, Burnier MN. The Phenotypical Characterization of Dual-Nature Hybrid Cells in Uveal Melanoma. Cancers (Basel) 2024; 16:3231. [PMID: 39335202 PMCID: PMC11429545 DOI: 10.3390/cancers16183231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Metastasis, occurring years after primary diagnosis, represents a poor prognosis in uveal melanoma (UM)-affected individuals. The nature of cells involved in this process is under debate. Circulating hybrid cells that have combined tumor and immune cell features found in blood were predictive of metastasis and may correspond to dual-nature cells (DNC) in the primary tumor. Herein, we sought to determine the presence of DNCs in primary UM tumors, the cell types involved in their genesis, and their ability to be formed in vitro. METHODS UM lesions (n = 38) were immunolabeled with HMB45 in combination with immune-cell-specific antibodies. In parallel, we co-cultured UM cells and peripheral blood mononuclear cells (PBMCs) to analyze DNC formation. RESULTS HMB45+/CD45+ DNCs were present in 90% (26/29) of the tumors, HMB45+/CD8+ DNCs were present in 93% (26/28), and HMB45+/CD68+ DNCs were present in 71% (17/24). DNCs formed with CD8+ and CD68+ cells were positively correlated to the infiltration of their respective immune cells. Notably, UM cells were prone to hybridize with PBMCs in vitro. CONCLUSIONS This phenotypical characterization of DNCs in UM demonstrates that CD8+ T-cells and macrophages are capable of DNC formation, and they are important for better understanding metastatic dissemination, thus paving the path towards novel therapeutic avenues.
Collapse
Affiliation(s)
- Emily Marcotte
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Alicia Goyeneche
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Mohamed Abdouh
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Julia Valdemarin Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H3A 3T2, Canada
| | - Miguel Noel Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
3
|
Patel RK, Parappilly MS, Walker BS, Heussner RT, Fung A, Chang YH, Kardosh A, Lopez CD, Mayo SC, Wong MH. Exploratory Analyses of Circulating Neoplastic-Immune Hybrid Cells as Prognostic Biomarkers in Advanced Intrahepatic Cholangiocarcinoma. Int J Mol Sci 2024; 25:9198. [PMID: 39273147 PMCID: PMC11395231 DOI: 10.3390/ijms25179198] [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: 07/29/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Existing clinical biomarkers do not reliably predict treatment response or disease progression in patients with advanced intrahepatic cholangiocarcinoma (ICC). Circulating neoplastic-immune hybrid cells (CHCs) have great promise as a blood-based biomarker for patients with advanced ICC. Peripheral blood specimens were longitudinally collected from patients with advanced ICC enrolled in the HELIX-1 phase II clinical trial (NCT04251715). CHCs were identified by co-expression of pan-cytokeratin (CK) and CD45, and levels were correlated to patient clinical disease course. Unsupervised machine learning was then performed to extract their morphological features to compare them across disease courses. Five patients were included in this study, with a median of nine specimens collected per patient. A median of 13.5 CHCs per 50,000 peripheral blood mononuclear cells were identified at baseline, and levels decreased to zero following the initiation of treatment in all patients. Counts remained undetectable in three patients who demonstrated end-of-trial clinical treatment response and conversely increased in two patients with evidence of therapeutic resistance. In the post-trial surveillance period, interval counts increased prior to or at the time of clinical progression in three patients and remain undetectable in one patient with continued long-term disease stability. Using our machine learning platform, treatment-resistant CHCs exhibited upregulation of CK and downregulation of CD45 relative to treatment-responsive CHCs. CHCs represent a promising blood-based biomarker to supplement traditional radiographic and biochemical measures.
Collapse
Affiliation(s)
- Ranish K. Patel
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Michael S. Parappilly
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Brett S. Walker
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Robert T. Heussner
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Alice Fung
- Department of Diagnostic Radiology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Adel Kardosh
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Charles D. Lopez
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Skye C. Mayo
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| | - Melissa H. Wong
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97201, USA
| |
Collapse
|
4
|
Kulbay M, Marcotte E, Remtulla R, Lau THA, Paez-Escamilla M, Wu KY, Burnier MN. Uveal Melanoma: Comprehensive Review of Its Pathophysiology, Diagnosis, Treatment, and Future Perspectives. Biomedicines 2024; 12:1758. [PMID: 39200222 PMCID: PMC11352094 DOI: 10.3390/biomedicines12081758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/02/2024] Open
Abstract
Uveal melanoma (UM) is the most common intraocular malignancy in adults. Recent advances highlight the role of tumor-derived extracellular vesicles (TEV) and circulating hybrid cells (CHC) in UM tumorigenesis. Bridged with liquid biopsies, a novel technology that has shown incredible performance in detecting cancer cells or products derived from tumors in bodily fluids, it can significantly impact disease management and outcome. The aim of this comprehensive literature review is to provide a summary of current knowledge and ongoing advances in posterior UM pathophysiology, diagnosis, and treatment. The first section of the manuscript discusses the complex and intricate role of TEVs and CHCs. The second part of this review delves into the epidemiology, etiology and risk factors, clinical presentation, and prognosis of UM. Third, current diagnostic methods, ensued by novel diagnostic tools for the early detection of UM, such as liquid biopsies and artificial intelligence-based technologies, are of paramount importance in this review. The fundamental principles, limits, and challenges associated with these diagnostic tools, as well as their potential as a tracker for disease progression, are discussed. Finally, a summary of current treatment modalities is provided, followed by an overview of ongoing preclinical and clinical research studies to provide further insights on potential biomolecular pathway alterations and therapeutic targets for the management of UM. This review is thus an important resource for all healthcare professionals, clinicians, and researchers working in the field of ocular oncology.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Emily Marcotte
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Raheem Remtulla
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Tsz Hin Alexander Lau
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Manuel Paez-Escamilla
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
| | - Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada;
| | - Miguel N. Burnier
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada; (M.K.); (R.R.); (T.H.A.L.); (M.P.-E.)
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
5
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Pang A, Farley HC, Gillingham AR, Dawson AD, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Detection of neoplastic-immune hybrid cells with metastatic properties in uveal melanoma. Biomark Res 2024; 12:67. [PMID: 39030653 PMCID: PMC11264923 DOI: 10.1186/s40364-024-00609-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/18/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Uveal melanoma is the most common non-cutaneous melanoma and is an intraocular malignancy affecting nearly 7,000 individuals per year worldwide. Of these, approximately 50% will progress to metastatic disease for which there are currently no effective curative therapies. Despite advances in molecular profiling and metastatic stratification of uveal melanoma tumors, little is known regarding their underlying biology of metastasis. Our group has identified a disseminated neoplastic cell population characterized by co-expression of immune and melanoma proteins, circulating hybrid cells (hybrids), in patients with uveal melanoma. Compared to circulating tumor cells, which lack expression of immune proteins, hybrids are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. METHODS To ascertain mechanisms underlying enhanced hybrid cell dissemination we identified hybrid cells within primary uveal melanoma tumors using single cell RNA sequencing (n = 8) and evaluated their gene expression and predicted ligand-receptor interactions in relation to other melanoma and immune cells within the primary tumor. We then verified expression of upregulated hybrid pathways within patient-matched tumor and peripheral blood hybrids (n = 4) using cyclic immunofluorescence and quantified their protein expression relative to other non-hybrid tumor and disseminated tumor cells. RESULTS Among the top upregulated genes and pathways in hybrid cells were those involved in enhanced cell motility and cytoskeletal rearrangement, immune evasion, and altered cellular metabolism. In patient-matched tumor and peripheral blood, we verified gene expression by examining concordant protein expression for each pathway category: TMSB10 (cell motility), CD74 (immune evasion) and GPX1 (metabolism). Both TMSB10 and GPX1 were expressed on significantly higher numbers of disseminated hybrid cells compared to circulating tumor cells, and CD74 and GPX1 were expressed on more disseminated hybrids than tumor-resident hybrids. Lastly, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting metastasis including GAS6-AXL, CXCL12-CXCR4, LGALS9-P4HB and IGF1-IGFR1. CONCLUSION These findings highlight the importance of TMSB10, GPX1 and CD74 for successful hybrid cell dissemination and survival in circulation. Our results contribute to the understanding of uveal melanoma tumor progression and interactions between tumor cells and immune cells in the tumor microenvironment that may promote metastasis.
Collapse
Affiliation(s)
- Ashley N Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Patrick Conley
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Christopher D Klocke
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Sidharth K Sengupta
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Amara Pang
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Hannah C Farley
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Abigail R Gillingham
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Aubrey D Dawson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Yichen Fan
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Alison H Skalet
- Casey Eye Institute, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Guanming Wu
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA.
- Knight Cancer Institute, OHSU, Portland, OR, USA.
| |
Collapse
|
6
|
Fuentes-Rodriguez A, Mitchell A, Guérin SL, Landreville S. Recent Advances in Molecular and Genetic Research on Uveal Melanoma. Cells 2024; 13:1023. [PMID: 38920653 PMCID: PMC11201764 DOI: 10.3390/cells13121023] [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: 04/10/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Uveal melanoma (UM), a distinct subtype of melanoma, presents unique challenges in its clinical management due to its complex molecular landscape and tendency for liver metastasis. This review highlights recent advancements in understanding the molecular pathogenesis, genetic alterations, and immune microenvironment of UM, with a focus on pivotal genes, such as GNAQ/11, BAP1, and CYSLTR2, and delves into the distinctive genetic and chromosomal classifications of UM, emphasizing the role of mutations and chromosomal rearrangements in disease progression and metastatic risk. Novel diagnostic biomarkers, including circulating tumor cells, DNA and extracellular vesicles, are discussed, offering potential non-invasive approaches for early detection and monitoring. It also explores emerging prognostic markers and their implications for patient stratification and personalized treatment strategies. Therapeutic approaches, including histone deacetylase inhibitors, MAPK pathway inhibitors, and emerging trends and concepts like CAR T-cell therapy, are evaluated for their efficacy in UM treatment. This review identifies challenges in UM research, such as the limited treatment options for metastatic UM and the need for improved prognostic tools, and suggests future directions, including the discovery of novel therapeutic targets, immunotherapeutic strategies, and advanced drug delivery systems. The review concludes by emphasizing the importance of continued research and innovation in addressing the unique challenges of UM to improve patient outcomes and develop more effective treatment strategies.
Collapse
Affiliation(s)
- Aurélie Fuentes-Rodriguez
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Andrew Mitchell
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Sylvain L. Guérin
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Solange Landreville
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| |
Collapse
|
7
|
Whalen RM, Anderson AN, Jones JA, Sims Z, Chang YH, Nederlof MA, Wong MH, Gibbs SL. Ultra high content analyses of circulating and tumor associated hybrid cells reveal phenotypic heterogeneity. Sci Rep 2024; 14:7350. [PMID: 38538742 PMCID: PMC10973471 DOI: 10.1038/s41598-024-57381-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/18/2024] [Indexed: 07/03/2024] Open
Abstract
Persistently high, worldwide mortality from cancer highlights the unresolved challenges of disease surveillance and detection that impact survival. Development of a non-invasive, blood-based biomarker would transform survival from cancer. We demonstrate the functionality of ultra-high content analyses of a newly identified population of tumor cells that are hybrids between neoplastic and immune cells in patient matched tumor and peripheral blood specimens. Using oligonucleotide conjugated antibodies (Ab-oligo) permitting cyclic immunofluorescence (cyCIF), we present analyses of phenotypes among tumor and peripheral blood hybrid cells. Interestingly, the majority of circulating hybrid cell (CHC) subpopulations were not identified in tumor-associated hybrids. These results highlight the efficacy of ultra-high content phenotypic analyses using Ab-oligo based cyCIF applied to both tumor and peripheral blood specimens. The combination of a multiplex phenotypic profiling platform that is gentle enough to analyze blood to detect and evaluate disseminated tumor cells represents a novel approach to exploring novel tumor biology and potential utility for developing the population as a blood-based biomarker in cancer.
Collapse
Affiliation(s)
- Riley M Whalen
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA
| | - Ashley N Anderson
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
| | - Zachary Sims
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | | | - Melissa H Wong
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA.
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA.
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.
| |
Collapse
|
8
|
Magri V, De Renzi G, Marino L, De Meo M, Siringo M, Gelibter A, Gareri R, Cataldi C, Giannini G, Santini D, Nicolazzo C, Gazzaniga P. Circulating Cancer-Associated Macrophage-like Cells as a Blood-Based Biomarker of Response to Immune Checkpoint Inhibitors. Int J Mol Sci 2024; 25:3752. [PMID: 38612563 PMCID: PMC11011814 DOI: 10.3390/ijms25073752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Evidence has been provided that circulating cancer-associated macrophage-like cell (CAM-L) numbers increase in response to chemotherapy, with an inverse trend compared to circulating tumor cells (CTCs). In the era of evolving cancer immunotherapy, whether CAM-Ls might have a potential role as predictive biomarkers of response has been unexplored. We evaluated whether a serial blood evaluation of CTC to CAM-L ratio might predict response to immune checkpoint inhibitors in a cohort of non-small-cell lung cancer patients. At baseline, CTCs, CAM-Ls, and the CTC/CAM-L ratio significantly correlate with both progression-free survival (PFS) and overall survival (OS). The baseline CTC/CAM-L ratio was significantly different in early progressors (4.28 ± 3.21) compared to long responders (0.42 ± 0.47) (p = 0.001). In patients treated with immune checkpoint inhibitors, a CTC/CAM-L ratio ≤ 0.25 at baseline is associated with better PFS and OS. A baseline CTC/CAM-L ratio ≤ 0.25 is statistically significant to discriminate early progressions from durable response. The results of the present pilot study suggest that CAM-Ls together with CTCs could play an important role in evaluating patients treated with cancer immunotherapy.
Collapse
Affiliation(s)
- Valentina Magri
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.G.); (C.C.); (D.S.)
| | - Gianluigi De Renzi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (G.G.); (C.N.); (P.G.)
| | - Luca Marino
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy;
| | - Michela De Meo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (G.G.); (C.N.); (P.G.)
| | - Marco Siringo
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.G.); (C.C.); (D.S.)
| | - Alain Gelibter
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.G.); (C.C.); (D.S.)
| | - Roberta Gareri
- UOC di Oncologia Medica, Ospedale Leopoldo Parodi Delfino, 00034 Colleferro, Italy;
| | - Chiara Cataldi
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.G.); (C.C.); (D.S.)
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (G.G.); (C.N.); (P.G.)
| | - Daniele Santini
- Department of Pathology, Oncology and Radiology, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.G.); (C.C.); (D.S.)
| | - Chiara Nicolazzo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (G.G.); (C.N.); (P.G.)
| | - Paola Gazzaniga
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.D.R.); (M.D.M.); (G.G.); (C.N.); (P.G.)
| |
Collapse
|
9
|
Patel RK, Parappilly M, Rahman S, Schwantes IR, Sewell M, Giske NR, Whalen RM, Durmus NG, Wong MH. The Hallmarks of Circulating Hybrid Cells. Results Probl Cell Differ 2024; 71:467-485. [PMID: 37996690 DOI: 10.1007/978-3-031-37936-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
While tumor metastases represent the primary driver of cancer-related mortality, our understanding of the mechanisms that underlie metastatic initiation and progression remains incomplete. Recent work identified a novel tumor-macrophage hybrid cell population, generated through the fusion between neoplastic and immune cells. These hybrid cells are detected in primary tumor tissue, peripheral blood, and in metastatic sites. In-depth analyses of hybrid cell biology indicate that they can exploit phenotypic properties of both parental tumor and immune cells, in order to intravasate into circulation, evade the immune response, and seed tumors at distant sites. Thus, it has become increasingly evident that the development and dissemination of tumor-immune hybrid cells play an intricate and fundamental role in the metastatic cascade and can provide invaluable information regarding tumor characteristics and patient prognostication. In this chapter, we review the current understanding of this novel hybrid cell population, the specific hallmarks of cancer that these cells exploit to promote cancer progression and metastasis, and discuss exciting new frontiers that remain to be explored.
Collapse
Affiliation(s)
- Ranish K Patel
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Michael Parappilly
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Shahrose Rahman
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Issac R Schwantes
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Marisa Sewell
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Nicole R Giske
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Riley M Whalen
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Naside Gozde Durmus
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Sieler M, Dittmar T. Cell Fusion and Syncytia Formation in Cancer. Results Probl Cell Differ 2024; 71:433-465. [PMID: 37996689 DOI: 10.1007/978-3-031-37936-9_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
The natural phenomenon of cell-cell fusion does not only take place in physiological processes, such as placentation, myogenesis, or osteoclastogenesis, but also in pathophysiological processes, such as cancer. More than a century ago postulated, today the hypothesis that the fusion of cancer cells with normal cells leads to the formation of cancer hybrid cells with altered properties is in scientific consensus. Some studies that have investigated the mechanisms and conditions for the fusion of cancer cells with other cells, as well as studies that have characterized the resulting cancer hybrid cells, are presented in this review. Hypoxia and the cytokine TNFα, for example, have been found to promote cell fusion. In addition, it has been found that both the protein Syncytin-1, which normally plays a role in placentation, and phosphatidylserine signaling on the cell membrane are involved in the fusion of cancer cells with other cells. In human cancer, cancer hybrid cells were detected not only in the primary tumor, but also in the circulation of patients as so-called circulating hybrid cells, where they often correlated with a worse outcome. Although some data are available, the questions of how and especially why cancer cells fuse with other cells are still not fully answered.
Collapse
Affiliation(s)
- Mareike Sieler
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Witten, Germany.
| | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Witten, Germany
| |
Collapse
|
11
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Pang A, Farley HC, Gillingham AR, Dawson AD, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Detection of neoplastic-immune hybrid cells with metastatic properties in uveal melanoma. RESEARCH SQUARE 2023:rs.3.rs-3694879. [PMID: 38106024 PMCID: PMC10723549 DOI: 10.21203/rs.3.rs-3694879/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Uveal melanoma is the most common non-cutaneous melanoma and is an intraocular malignancy affecting nearly 7,000 individuals per year worldwide. Of these, approximately 50% will progress to metastatic disease for which there are currently no effective therapies. Despite advances in molecular profiling and metastatic stratification of uveal melanoma tumors, little is known regarding their underlying biology of metastasis. Our group has identified a disseminated neoplastic cell population characterized by co-expression of immune and melanoma proteins, circulating hybrid cells (hybrids), in patients with uveal melanoma. Compared to circulating tumor cells, which lack expression of immune proteins, hybrids are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. Methods To ascertain mechanisms underlying enhanced hybrid cell dissemination we identified hybrid cells within primary uveal melanoma tumors using single cell RNA sequencing and evaluated their gene expression and predicted ligand-receptor interactions in relation to other melanoma and immune cells within the primary tumor. We then verified expression of upregulated hybrid pathways within patient-matched tumor and peripheral blood hybrids using cyclic immunofluorescence and quantified their protein expression relative to other non-hybrid tumor and disseminated tumor cells. Results Among the top upregulated genes and pathways in hybrid cells were those involved in enhanced cell motility and cytoskeletal rearrangement, immune evasion, and altered cellular metabolism. In patient-matched tumor and peripheral blood, we verified gene expression by examining concordant protein expression for each pathway category: TMSB10 (cell motility), CD74 (immune evasion) and GPX1 (metabolism). Both TMSB10 and GPX1 were expressed on significantly higher numbers of disseminated hybrid cells compared to circulating tumor cells, and CD74 and GPX1 were expressed on more disseminated hybrids than tumor-resident hybrids. Lastly, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting metastasis including GAS6-AXL, CXCL12-CXCR4, LGALS9-P4HB and IGF1-IGFR1. Conclusion These findings highlight the importance of TMSB10, GPX1 and CD74 for successful hybrid cell dissemination and survival in circulation. Our results contribute to the understanding of uveal melanoma tumor progression and interactions between tumor cells and immune cells in the tumor microenvironment that may promote metastasis.
Collapse
|
12
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Robinson TL, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Analysis of uveal melanoma scRNA sequencing data identifies neoplastic-immune hybrid cells that exhibit metastatic potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563815. [PMID: 37961378 PMCID: PMC10634980 DOI: 10.1101/2023.10.24.563815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Uveal melanoma (UM) is the most common non-cutaneous melanoma and is an intraocular malignancy that affects nearly 7,000 individuals per year worldwide. Of these, nearly 50% will progress to metastatic disease for which there are currently no effective therapies. Despite advances in the molecular profiling and metastatic stratification of class 1 and 2 UM tumors, little is known regarding the underlying biology of UM metastasis. Our group has identified a disseminated tumor cell population characterized by co-expression of immune and melanoma proteins, (circulating hybrid cells (CHCs), in patients with UM. Compared to circulating tumor cells, CHCs are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. To identify mechanisms underlying enhanced hybrid cell dissemination we sought to identify hybrid cells within a primary UM single cell RNA-seq dataset. Using rigorous doublet discrimination approaches, we identified UM hybrids and evaluated their gene expression, predicted ligand-receptor status, and cell-cell communication state in relation to other melanoma and immune cells within the primary tumor. We identified several genes and pathways upregulated in hybrid cells, including those involved in enhancing cell motility and cytoskeleton rearrangement, evading immune detection, and altering cellular metabolism. In addition, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting cancer metastasis including IGF1-IGFR1, GAS6-AXL, LGALS9-P4HB, APP-CD74 and CXCL12-CXCR4. These results contribute to our understanding of tumor progression and interactions between tumor cells and immune cells in the UM microenvironment that may promote metastasis.
Collapse
|
13
|
Patel RK, Rahman S, Schwantes IR, Bartlett A, Eil R, Farsad K, Fowler K, Goodyear SM, Hansen L, Kardosh A, Nabavizadeh N, Rocha FG, Tsikitis VL, Wong MH, Mayo SC. Updated Management of Colorectal Cancer Liver Metastases: Scientific Advances Driving Modern Therapeutic Innovations. Cell Mol Gastroenterol Hepatol 2023; 16:881-894. [PMID: 37678799 PMCID: PMC10598050 DOI: 10.1016/j.jcmgh.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths in the United States and accounts for an estimated 1 million deaths annually worldwide. The liver is the most common site of metastatic spread from colorectal cancer, significantly driving both morbidity and mortality. Although remarkable advances have been made in recent years in the management for patients with colorectal cancer liver metastases, significant challenges remain in early detection, prevention of progression and recurrence, and in the development of more effective therapeutics. In 2017, our group held a multidisciplinary state-of-the-science symposium to discuss the rapidly evolving clinical and scientific advances in the field of colorectal liver metastases, including novel early detection and prognostic liquid biomarkers, identification of high-risk cohorts, advances in tumor-immune therapy, and different regional and systemic therapeutic strategies. Since that time, there have been scientific discoveries translating into therapeutic innovations addressing the current management challenges. These innovations are currently reshaping the treatment paradigms and spurring further scientific discovery. Herein, we present an updated discussion of both the scientific and clinical advances and future directions in the management of colorectal liver metastases, including adoptive T-cell therapies, novel blood-based biomarkers, and the role of the tumor microbiome. In addition, we provide a comprehensive overview detailing the role of modern multidisciplinary clinical approaches used in the management of patients with colorectal liver metastases, including considerations toward specific molecular tumor profiles identified on next generation sequencing, as well as quality of life implications for these innovative treatments.
Collapse
Affiliation(s)
- Ranish K Patel
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Shahrose Rahman
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Issac R Schwantes
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Alexandra Bartlett
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon
| | - Robert Eil
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon; The Knight Cancer Institute, OHSU, Portland, Oregon
| | - Khashayar Farsad
- Charles T. Dotter Department of Interventional Radiology, OHSU, Portland, Oregon
| | - Kathryn Fowler
- Department of Surgery, Oregon Health & Science University (OHSU), Portland, Oregon
| | - Shaun M Goodyear
- The Knight Cancer Institute, OHSU, Portland, Oregon; Division of Hematology and Oncology, School of Medicine, OHSU, Portland, Oregon
| | - Lissi Hansen
- The Knight Cancer Institute, OHSU, Portland, Oregon; School of Nursing, OHSU, Portland, Oregon
| | - Adel Kardosh
- The Knight Cancer Institute, OHSU, Portland, Oregon; Division of Hematology and Oncology, School of Medicine, OHSU, Portland, Oregon
| | - Nima Nabavizadeh
- The Knight Cancer Institute, OHSU, Portland, Oregon; Department of Radiation Medicine, OHSU, Portland, Oregon
| | - Flavio G Rocha
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon; The Knight Cancer Institute, OHSU, Portland, Oregon
| | - V Liana Tsikitis
- The Knight Cancer Institute, OHSU, Portland, Oregon; Division of Gastrointestinal Surgery, Department of Surgery, OHSU, Portland, Oregon
| | - Melissa H Wong
- The Knight Cancer Institute, OHSU, Portland, Oregon; Department of Cell, Developmental and Cancer Biology, OHSU, Portland, Oregon
| | - Skye C Mayo
- Division of Surgical Oncology, Department of Surgery, OHSU, Portland, Oregon; The Knight Cancer Institute, OHSU, Portland, Oregon.
| |
Collapse
|
14
|
Heussner RT, Whalen RM, Anderson A, Theison H, Baik J, Gibbs S, Wong MH, Chang YH. Quantitative image analysis pipeline for detecting circulating hybrid cells in immunofluorescence images with human-level accuracy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554733. [PMID: 37662330 PMCID: PMC10473764 DOI: 10.1101/2023.08.24.554733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Circulating hybrid cells (CHCs) are a newly discovered, tumor-derived cell population identified in the peripheral blood of cancer patients and are thought to contribute to tumor metastasis. However, identifying CHCs by immunofluorescence (IF) imaging of patient peripheral blood mononuclear cells (PBMCs) is a time-consuming and subjective process that currently relies on manual annotation by laboratory technicians. Additionally, while IF is relatively easy to apply to tissue sections, its application on PBMC smears presents challenges due to the presence of biological and technical artifacts. To address these challenges, we present a robust image analysis pipeline to automate the detection and analyses of CHCs in IF images. The pipeline incorporates quality control to optimize specimen preparation protocols and remove unwanted artifacts, leverages a β-variational autoencoder (VAE) to learn meaningful latent representations of single-cell images and employs a support vector machine (SVM) classifier to achieve human-level CHC detection. We created a rigorously labeled IF CHC dataset including 9 patients and 2 disease sites with the assistance of 10 annotators to evaluate the pipeline. We examined annotator variation and bias in CHC detection and then provided guidelines to optimize the accuracy of CHC annotation. We found that all annotators agreed on CHC identification for only 65% of the cells in the dataset and had a tendency to underestimate CHC counts for regions of interest (ROI) containing relatively large amounts of cells (>50,000) when using conventional enumeration methods. On the other hand, our proposed approach is unbiased to ROI size. The SVM classifier trained on the β-VAE encodings achieved an F1 score of 0.80, matching the average performance of annotators. Our pipeline enables researchers to explore the role of CHCs in cancer progression and assess their potential as a clinical biomarker for metastasis. Further, we demonstrate that the pipeline can identify discrete cellular phenotypes among PBMCs, highlighting its utility beyond CHCs.
Collapse
Affiliation(s)
- Robert T. Heussner
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97201, USA
| | - Riley M. Whalen
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Ashley Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Heather Theison
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Joseph Baik
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97201, USA
| | - Summer Gibbs
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
15
|
Zhang C, Xiao J, Fa L, Jiang F, Jiang H, Zhou L, Xu Z. Identification of co-expressed gene networks promoting CD8 + T cell infiltration and having prognostic value in uveal melanoma. BMC Ophthalmol 2023; 23:354. [PMID: 37563735 PMCID: PMC10416479 DOI: 10.1186/s12886-023-03098-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Current immunotherapies are unsatisfactory against uveal melanoma (UM); however, elevated CD8+ T cell infiltration level indicates poor prognosis in UM. Here, we aimed to identify co-expressed gene networks promoting CD8+ T cell infiltration in UM and created a prognostic hazard model based on the identified hub genes. Raw data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Stromal-immune comprehensive score (ESTIMATE) was used to evaluate the immune-infiltration landscape of the tumor microenvironment. Single-Sample Gene Set Enrichment Analysis (ssGSEA) and Weighted Correlation Network Analysis (WGCNA) were used to quantify CD8+ T cell infiltration level and identify hub genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to analyze the biological processes. Least absolute shrinkage and selection operator (LASSO) Cox regression were used to establish a prognostic model, which was further validated. Finally, pan-cancer analysis evaluated these genes to be associated with CD8+ T cell infiltration in other tumors. In conclusion, the proposed four-gene (PTPN12, IDH2, P2RX4, and KDELR2) prognostic hazard model had satisfactory prognostic ability. These hub genes may promote CD8+ T cell infiltration in UM through antigen presentation, and CD8+ T cell possibly function as Treg, resulting in poor prognosis. These findings might facilitate the development of novel immunotherapies.
Collapse
Affiliation(s)
- Chun Zhang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Jing Xiao
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Luzhong Fa
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Fanwen Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Hui Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Lin Zhou
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Zhuping Xu
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China.
| |
Collapse
|