1
|
Meliante PG, Pizzolante S, Perna L, Filippi C, Bandiera G, Barbato C, Minni A, de Vincentiis M, Covelli E. Molecular Markers of Occult Lymph Node Metastasis in Head and Neck Squamous Cell Carcinoma (HNSCC) Patients. FRONT BIOSCI-LANDMRK 2025; 30:25267. [PMID: 40018925 DOI: 10.31083/fbl25267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 03/01/2025]
Abstract
The accurate diagnosis of regional lymph node metastasis is critical for guiding treatment decisions in head and neck cancer patients. Despite advances in imaging techniques, detecting nodal metastasis using radiology remains challenging, leading to potential undertreatment or overtreatment. This review aims to identify molecular markers associated with occult metastasis in head and neck squamous cell carcinoma (HNSCC) patients. We divided the results by subsite for markers: lymph node analysis (microRNAs, myosin-5a (MYO5A), ring finger protein 145 (RNF145), F-box only protein 32 (FBXO32), CTONG2002744, cytokeratin 14 (CK14), eukaryotic initiation factor 4E (eIF4E), desmoglein-3 (DSG3), microsatellite D9S171, squamous cell carcinoma antigen, cytokeratin, tumor budding score, human papillomavirus-DNA (HPV-DNA), tumor infiltrating lymphocytes, sentinel lymph node analysis techniques, single fiber reflectance spectroscopy, radiological techniques), tumor tissue analysis (activin A, carcinoma-associated fibroblasts, cyclins, β-catenin, histopathology, genetic amplifications, DNA methylation, ecotropic viral integration site 1, CC-chemokine receptor 7, melanoma associated-A antigens, vascular endothelial growth factor-C (VEGF-C), panitumumab, epidermal growth factor receptor (EGFR), cornulin, total protein analysis, CD133, NANOG homeobox, neurogenic locus notch homolog protein 1 (NOTCH1), metastasis-associated protein 1, 14-3-3-zeta, E-cadherin, focal adhesion kinase, p-epithelial-mesenchymal transition (EMT), small proline rich protein 1B (SPRR1B), transcription factor NKX3-1, DNA copy number aberrations, microfibril-associated protein 5 (MFAP5), troponin C1, slow skeletal and cardiac type (TNNC1), matrix Gla protein (MGP), fibroblast growth factor binding protein 1 (FBFBP1), F-box protein 32 (FBXO32), fatty acid binding protein 5, B cell-specific Moloney murine leukemia virus integration site 1, podoplanin, p53, Bcl-2, epidermal growth factor receptor (EGFR), Ki67, cyclin D1, cox-2, semaphorin-3F, neuropilin-2, histologic features, cellular dissociation grade, prospero homeobox protein 1, radiologic features, Ki-67, poly (ADP-ribose) polymerase (PARP), Bcl-2 associated agonist of cell death (BAD), caspase-9, vascular endothelial growth factor A (VEGF-A), HPV, p16, methylation status of long interspersed element 1 (LINE-1) and Alu elements, mesenchymal-epithelial transition (MET), gene expression analyses, molecular subtypes) and blood markers (standard blood analysis indexes and ratios, circulating tumor cells, HPV-DNA, CD-31, bone marrow analysis). Several promising markers were identified, including miR-205, desmoglein 3 (DSG3), pan-cytokeratin (CK) AE1/AE3, HPV-16, activin-A, cyclin D1, E-cadherin, and neural progenitor lineage (NPL) that demonstrated effectiveness across multiple studies. Future research should focus on exploring combination scoring systems to improve diagnostic precision and optimize treatment selection in HNSCC patients.
Collapse
Affiliation(s)
- Piero Giuseppe Meliante
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "Sapienza" University of Rome, Sant'Andrea Hospital, 00189 Rome, Italy
| | - Sofia Pizzolante
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "Sapienza" University of Rome, Sant'Andrea Hospital, 00189 Rome, Italy
| | - Luca Perna
- Department of Otolaryngology, San Leonardo Hospital, 80053 Castellammare di Stabia, Italy
| | - Chiara Filippi
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "Sapienza" University of Rome, Sant'Andrea Hospital, 00189 Rome, Italy
| | - Giorgio Bandiera
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "Sapienza" University of Rome, Sant'Andrea Hospital, 00189 Rome, Italy
| | - Christian Barbato
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Sapienza University of Rome, 00161 Roma, Italy
| | - Antonio Minni
- Department of Sense Organs, Sapienza University of Rome, 00161 Roma, Italy
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, Azienda Sanitaria Locale (ASL) Rieti-Sapienza University, 02100 Rieti, Italy
| | | | - Edoardo Covelli
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "Sapienza" University of Rome, Sant'Andrea Hospital, 00189 Rome, Italy
| |
Collapse
|
2
|
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
|
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
|
Zhang C, Yang H, Chang C, Li R, Xiong J, Kang D, He D, Liu XJ, Cheng K, Cao D. Serplulimab combined with gemcitabine, nab-paclitaxel and stereotactic body radiotherapy as the first-line treatment for patients with metastatic pancreatic adenocarcinoma in China: a multicentre, single-arm, phase II trial (ICSBR) protocol. BMJ Open 2024; 14:e084274. [PMID: 39013651 PMCID: PMC11253733 DOI: 10.1136/bmjopen-2024-084274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
INTRODUCTION Patients with pancreatic ductal adenocarcinoma (PDAC) remain a poor prognosis despite the development of chemotherapy. Although programmed cell death 1 (PD-1) blockade has shown great efficacy in various solid tumours, its application in treating PDAC is limited. Recent studies have indicated that chemotherapy or stereotactic body radiotherapy (SBRT) may improve the antitumour effect of PD-1 blockade in patients with PDAC. The objective of this study is to evaluate the efficacy and safety of combined therapy comprising PD-1 blockade, gemcitabine plus nab-paclitaxel chemotherapy and SBRT for patients with metastatic PDAC. METHODS AND ANALYSIS This is a multicentre, single-arm, prospective phase II clinical trial. Forty-three patients diagnosed with metastatic PDAC will be enrolled. The eligible patients will be intravenously administered 1000 mg/m2 gemcitabine and 125 mg/m2 nab-paclitaxel on days 1 and 8 of the 21-day cycle. Serplulimab (200 mg) will be administered intravenously on day 1 of the 21-day cycle. Furthermore, during the second cycle, the patients will undergo SBRT with doses of 33 Gy in five fractions for primary lesions or doses of 24 Gy in three fractions for metastases. The primary endpoint is the 6-month progression-free survival (PFS) rate. The secondary endpoints overall survival, PFS, overall response rate, disease control rate, time to progression, duration of response, duration of disease control and safety. Moreover, this trial seeks to investigate biomarkers such as circulating tumour DNA and circulating hybrid cells in patients diagnosed with metastatic PDAC. ETHICS AND DISSEMINATION The study was approved by the Ethics Committee on Biomedical Research, West China Hospital of Sichuan University. The study results will be presented at international conferences and published in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ChiCTR2300073237.
Collapse
Affiliation(s)
- Chenyan Zhang
- Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Heqi Yang
- Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chen Chang
- Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ruizhen Li
- Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junjie Xiong
- Department of General Surgery, Sichuan University, Chengdu, Sichuan, China
| | - Deying Kang
- West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Du He
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | | | - Ke Cheng
- Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Dan Cao
- Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
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. Cytometry A 2024; 105:345-355. [PMID: 38385578 PMCID: PMC11217923 DOI: 10.1002/cyto.a.24826] [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: 09/14/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Circulating hybrid cells (CHCs) are a newly discovered, tumor-derived cell population found 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 to 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 analysis 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 data set including nine patients and two disease sites with the assistance of 10 annotators to evaluate the pipeline. We examined annotator variation and bias in CHC detection and 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 data set and had a tendency to underestimate CHC counts for regions of interest (ROIs) containing relatively large amounts of cells (>50,000) when using the conventional enumeration method. On the other hand, our proposed approach is unbiased to ROI size. The SVM classifier trained on the β-VAE embeddings achieved an F1 score of 0.80, matching the average performance of human 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, Oregon, USA
| | - Riley M. Whalen
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Ashley Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Heather Theison
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph Baik
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Summer Gibbs
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Ali AM, Raza A. scRNAseq and High-Throughput Spatial Analysis of Tumor and Normal Microenvironment in Solid Tumors Reveal a Possible Origin of Circulating Tumor Hybrid Cells. Cancers (Basel) 2024; 16:1444. [PMID: 38611120 PMCID: PMC11010995 DOI: 10.3390/cancers16071444] [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: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Metastatic cancer is a leading cause of death in cancer patients worldwide. While circulating hybrid cells (CHCs) are implicated in metastatic spread, studies documenting their tissue origin remain sparse, with limited candidate approaches using one-two markers. Utilizing high-throughput single-cell and spatial transcriptomics, we identified tumor hybrid cells (THCs) co-expressing epithelial and macrophage markers and expressing a distinct transcriptome. Rarely, normal tissue showed these cells (NHCs), but their transcriptome was easily distinguishable from THCs. THCs with unique transcriptomes were observed in breast and colon cancers, suggesting this to be a generalizable phenomenon across cancer types. This study establishes a framework for HC identification in large datasets, providing compelling evidence for their tissue residence and offering comprehensive transcriptomic characterization. Furthermore, it sheds light on their differential function and identifies pathways that could explain their newly acquired invasive capabilities. THCs should be considered as potential therapeutic targets.
Collapse
Affiliation(s)
- Abdullah Mahmood Ali
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Edward P Evans MDS Center, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - Azra Raza
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Edward P Evans MDS Center, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| |
Collapse
|
7
|
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: 12] [Impact Index Per Article: 6.0] [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
|
8
|
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
|
9
|
Chantre-Justino M, Alves G, Delmonico L. Clinical applications of liquid biopsy in HPV‐negative and HPV‐positive head and neck squamous cell carcinoma: advances and challenges. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:533-552. [PMID: 36071985 PMCID: PMC9446158 DOI: 10.37349/etat.2022.00099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/22/2022] [Indexed: 12/02/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) represent the most common epithelial tumors that arise from mucosa of the oral cavity, pharynx, and larynx. The development of HNSCCs is usually associated with tobacco use, alcohol consumption, and human papillomavirus (HPV) infection. Most HNSCCs are diagnosed in advanced states, leading to a worse clinical outcome. Screening tests based on potential biomarkers associated with HNSCCs could improve this scenario. Liquid biopsy has emerged as a promising area of cancer investigation, offering a minimally invasive approach to track circulating biomarkers in body fluids that could potentially contribute to the diagnosis, predict prognosis, and monitor response to treatment. This review will discuss translational studies describing the clinical applications of liquid biopsy in HPV-negative and HPV-positive HNSCCs focused on circulating nucleic acids [cell-free DNA (cfDNA) and cell-free RNA (cfRNA)], circulating tumor cells (CTCs), and extracellular vesicles (EVs), which can be found in plasma, serum, and saliva.
Collapse
Affiliation(s)
- Mariana Chantre-Justino
- 1Research Division, National Institute of Traumatology and Orthopaedics (INTO), Rio de Janeiro 20940-070, Brazil 2Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Gilda Alves
- 2Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Lucas Delmonico
- 3Oncoclínicas Precision Medicine, Vila Nova Conceição, São Paulo 04513-020, Brazil
| |
Collapse
|
10
|
Sutton TL, Patel RK, Anderson AN, Bowden SG, Whalen R, Giske NR, Wong MH. Circulating Cells with Macrophage-like Characteristics in Cancer: The Importance of Circulating Neoplastic-Immune Hybrid Cells in Cancer. Cancers (Basel) 2022; 14:cancers14163871. [PMID: 36010865 PMCID: PMC9405966 DOI: 10.3390/cancers14163871] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary In cancer, disseminated neoplastic cells circulating in blood are a source of tumor DNA, RNA, and protein, which can be harnessed to diagnose, monitor, and better understand the biology of the tumor from which they are derived. Historically, circulating tumor cells (CTCs) have dominated this field of study. While CTCs are shed directly into circulation from a primary tumor, they remain relatively rare, particularly in early stages of disease, and thus are difficult to utilize as a reliable cancer biomarker. Neoplastic-immune hybrid cells represent a novel subpopulation of circulating cells that are more reliably attainable as compared to their CTC counterparts. Here, we review two recently identified circulating cell populations in cancer—cancer-associated macrophage-like cells and circulating hybrid cells—and discuss the future impact for the exciting area of disseminated hybrid cells. Abstract Cancer remains a significant cause of mortality in developed countries, due in part to difficulties in early detection, understanding disease biology, and assessing treatment response. If effectively harnessed, circulating biomarkers promise to fulfill these needs through non-invasive “liquid” biopsy. While tumors disseminate genetic material and cellular debris into circulation, identifying clinically relevant information from these analytes has proven difficult. In contrast, cell-based circulating biomarkers have multiple advantages, including a source for tumor DNA and protein, and as a cellular reflection of the evolving tumor. While circulating tumor cells (CTCs) have dominated the circulating cell biomarker field, their clinical utility beyond that of prognostication has remained elusive, due to their rarity. Recently, two novel populations of circulating tumor-immune hybrid cells in cancer have been characterized: cancer-associated macrophage-like cells (CAMLs) and circulating hybrid cells (CHCs). CAMLs are macrophage-like cells containing phagocytosed tumor material, while CHCs can result from cell fusion between cancer and immune cells and play a role in the metastatic cascade. Both are detected in higher numbers than CTCs in peripheral blood and demonstrate utility in prognostication and assessing treatment response. Additionally, both cell populations are heterogeneous in their genetic, transcriptomic, and proteomic signatures, and thus have the potential to inform on heterogeneity within tumors. Herein, we review the advances in this exciting field.
Collapse
Affiliation(s)
- Thomas L. Sutton
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ranish K. Patel
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ashley N. Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Stephen G. Bowden
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Riley Whalen
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Nicole R. Giske
- Department of Cell, Developmental and Cancer Biology, 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
- Correspondence: ; Tel.: +1-503-494-8749; Fax: +1-503-494-4253
| |
Collapse
|
11
|
Semaphorin-3F/Neuropilin-2 Transcriptional Expression as a Predictive Biomarker of Occult Lymph Node Metastases in HNSCC. Cancers (Basel) 2022; 14:cancers14092259. [PMID: 35565388 PMCID: PMC9100497 DOI: 10.3390/cancers14092259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 12/24/2022] Open
Abstract
The expression of the semaphorin-3F (SEMA3F) and neuropilin-2 (NRP2) is involved in the regulation of lymphangiogenesis. The present study analyzes the relationship between the transcriptional expression of the SEMA3F-NRP2 genes and the presence of occult lymph node metastases in patients with cN0 head and neck squamous cell carcinomas. We analyzed the transcriptional expression of SEMA3F and NRP2 in a cohort of 53 patients with cN0 squamous cell carcinoma treated with an elective neck dissection. Occult lymph node metastases were found in 37.7% of the patients. Patients with occult lymph node metastases (cN0/pN+) had significantly lower SEMA3F expression values than patients without lymph node involvement (cN0/pN0). Considering the expression of the SEMA3F-NRP2 genes, patients were classified into two groups according to the risk of occult nodal metastasis: Group 1 (n = 34), high SEMA3F/low NRP2 expression, with a low risk of occult nodal involvement (14.7% cN0/pN+); Group 2 (n = 19), low SEMA3F or high SEMA3F/high NRP2 expression, with a high risk of occult nodal involvement (78.9% cN0/pN+). Multivariate analysis showed that patients in Group 2 had a 26.2 higher risk of lymph node involvement than patients in Group 1. There was a significant relationship between the transcriptional expression values of the SEMA3F-NRP2 genes and the risk of occult nodal metastases.
Collapse
|
12
|
Mukherjee S, Ali AM, Murty VV, Raza A. Mutation in SF3B1 gene promotes formation of polyploid giant cells in Leukemia cells. Med Oncol 2022; 39:65. [PMID: 35478057 PMCID: PMC9046281 DOI: 10.1007/s12032-022-01652-9] [Citation(s) in RCA: 6] [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: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/02/2022]
Abstract
Giant cells with polyploidy, termed polyploid giant cells, have been observed during normal growth, development, and pathologic states, such as solid cancer progression and resistance to therapy. Functional studies of polyploidal giant cancer cells (PGCC) provided evidence that they arise when normal diploid cells are stressed, show stem cell-like properties, and give rise to tumors. In the present study, we report in K562 leukemia cell line that introduction of the hotspot K700E mutation in the gene SF3B1 using CRISPR/Cas9 method results in an increased frequency of multinucleated polyploid giant cells resistant to chemotherapeutic agent and serum starvation stress. These giant cells with higher ploidy are distinct from multinucleated megakaryocytes, are proliferative, and are characterized by increased accumulation of mitochondria. PGCC have been previously documented in solid tumors. This is the first report describing PGCCs in a cell line derived from a liquid cancer where increased frequency of PGCCs is linked to a specific genetic event. Since SF3B1 mutations are predominantly seen in MDS and other hematologic malignancies, our current findings will have significant clinical implications.
Collapse
Affiliation(s)
- Sanjay Mukherjee
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Abdullah Mahmood Ali
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Vundavalli V Murty
- Department of Pathology and Cell Biology, and Institute for Cancer Genetics, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Azra Raza
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA.
- MDS Center, Columbia University Irving Medical Center, 177 Fort Washington Avenue, Milstein Hospital Building, Room 6GN-435, New York, NY, 10032, USA.
| |
Collapse
|
13
|
Kaigorodova EV, Kozik AV, Zavaruev IS, Grishchenko MY. Hybrid/Atypical Forms of Circulating Tumor Cells: Current State of the Art. BIOCHEMISTRY (MOSCOW) 2022; 87:380-390. [PMID: 35527376 PMCID: PMC8993035 DOI: 10.1134/s0006297922040071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cancer is one of the most common diseases worldwide, and its treatment is associated with many challenges such as drug and radioresistance and formation of metastases. These difficulties are due to tumor heterogeneity, which has many causes. One may be the cell fusion, a process that is relevant to both physiological (e.g., wound healing) and pathophysiological (cancer and viral infection) processes. This literature review aimed to summarize the existing data on the hybrid/atypical forms of circulating cancer cells and their role in tumor progression. For that, the bioinformatics search in universal databases, such as PubMed, NCBI, and Google Scholar was conducted by using the keywords “hybrid cancer cells”, “cancer cell fusion”, etc. In this review the latest information related to the hybrid tumor cells, theories of their genesis, characteristics of different variants with data from our own researches are presented. Many aspects of the hybrid cell research are still in their infancy. However, with the level of knowledge already accumulated, circulating hybrids such as CAML and CHC could be considered as promising biomarkers of cancerous tumors, and even more as a new approach to cancer treatment.
Collapse
Affiliation(s)
- Evgeniya V Kaigorodova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
- Siberian State Medical University, Tomsk, 634050, Russia
| | - Alexey V Kozik
- Siberian State Medical University, Tomsk, 634050, Russia
| | | | | |
Collapse
|