1
|
Guo DZ, Zhang X, Zhang SQ, Zhang SY, Zhang XY, Yan JY, Dong SY, Zhu K, Yang XR, Fan J, Zhou J, Huang A. Single-cell tumor heterogeneity landscape of hepatocellular carcinoma: unraveling the pro-metastatic subtype and its interaction loop with fibroblasts. Mol Cancer 2024; 23:157. [PMID: 39095854 PMCID: PMC11295380 DOI: 10.1186/s12943-024-02062-3] [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: 02/01/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Tumor heterogeneity presents a formidable challenge in understanding the mechanisms driving tumor progression and metastasis. The heterogeneity of hepatocellular carcinoma (HCC) in cellular level is not clear. METHODS Integration analysis of single-cell RNA sequencing data and spatial transcriptomics data was performed. Multiple methods were applied to investigate the subtype of HCC tumor cells. The functional characteristics, translation factors, clinical implications and microenvironment associations of different subtypes of tumor cells were analyzed. The interaction of subtype and fibroblasts were analyzed. RESULTS We established a heterogeneity landscape of HCC malignant cells by integrated 52 single-cell RNA sequencing data and 5 spatial transcriptomics data. We identified three subtypes in tumor cells, including ARG1+ metabolism subtype (Metab-subtype), TOP2A+ proliferation phenotype (Prol-phenotype), and S100A6+ pro-metastatic subtype (EMT-subtype). Enrichment analysis found that the three subtypes harbored different features, that is metabolism, proliferating, and epithelial-mesenchymal transition. Trajectory analysis revealed that both Metab-subtype and EMT-subtype originated from the Prol-phenotype. Translation factor analysis found that EMT-subtype showed exclusive activation of SMAD3 and TGF-β signaling pathway. HCC dominated by EMT-subtype cells harbored an unfavorable prognosis and a deserted microenvironment. We uncovered a positive loop between tumor cells and fibroblasts mediated by SPP1-CD44 and CCN2/TGF-β-TGFBR1 interaction pairs. Inhibiting CCN2 disrupted the loop, mitigated the transformation to EMT-subtype, and suppressed metastasis. CONCLUSION By establishing a heterogeneity landscape of malignant cells, we identified a three-subtype classification in HCC. Among them, S100A6+ tumor cells play a crucial role in metastasis. Targeting the feedback loop between tumor cells and fibroblasts is a promising anti-metastatic strategy.
Collapse
Affiliation(s)
- De-Zhen Guo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Sen-Quan Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shi-Yu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiang-Yu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jia-Yan Yan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - San-Yuan Dong
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China
| | - Kai Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin-Rong Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| | - Ao Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Ferrell LD, Kakar S, Terracciano LM, Wee A. Tumours and Tumour-Like Lesions. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:842-946. [DOI: 10.1016/b978-0-7020-8228-3.00013-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Umetsu SE, Kakar S. Evaluating Liver Biopsies with Well-Differentiated Hepatocellular Lesions. Surg Pathol Clin 2023; 16:581-598. [PMID: 37536890 DOI: 10.1016/j.path.2023.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Needle core biopsies of liver lesions can be challenging, particularly in cases with limited material. The differential diagnosis for well-differentiated hepatocellular lesions includes focal nodular hyperplasia, hepatocellular adenoma, and well-differentiated hepatocellular carcinoma (HCC) in noncirrhotic liver, while dysplastic nodules and well-differentiated HCC are the primary considerations in cirrhotic liver. The first part of this review focuses on histochemical and immunohistochemical stains as well as molecular assays that are useful in the differential diagnosis. The second portion describes the features of hepatocellular adenoma subtypes and focuses on the differential diagnoses in commonly encountered clinicopathologic scenarios.
Collapse
Affiliation(s)
- Sarah E Umetsu
- Department of Pathology, University of California San Francisco, 505 Parnassus Avenue, Box 0102, San Francisco, CA 94143, USA.
| | - Sanjay Kakar
- Department of Pathology, University of California San Francisco, 505 Parnassus Avenue, Box 0102, San Francisco, CA 94143, USA
| |
Collapse
|
4
|
Kmeid M, Park YN, Chung T, Lukose G, Sullivan L, Brar R, Lee H. PSMA Immunohistochemistry in Hepatic Neoplasms: A Promising Diagnostic Marker With Potential Theranostic Applications. Am J Surg Pathol 2022; 46:1688-1699. [PMID: 36190927 DOI: 10.1097/pas.0000000000001971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Accurate classification of well-differentiated hepatocellular neoplasms can be challenging especially in core biopsies. Prostate-specific membrane antigen (PSMA) has been shown to highlight tumor-associated neovasculature in many nonprostatic solid tumors including hepatocellular carcinoma (HCC). Archived 164 hepatectomies and explants with 68 HCCs, 31 hepatocellular adenoma (HA), 24 dysplastic nodules (DN), and 42 metastases were retrieved, and pathologic parameters were evaluated. Sensitivity, specificity, accuracy, positive, and negative predictive values for correct diagnosis of HCC were calculated for PSMA and CD34 immunostains in tissue sections and HCC tissue microarrays. PSMA positivity was defined as capillarized sinusoidal/tumor-associated vessel staining involving ≥5% of the tumor area. In all, 55/68 (80.9%) HCC and 37/42 (88.1%) of liver metastasis were PSMA positive. PSMA was negative in HA, DN, and background liver (100% specificity). CD34 had a 98.5% sensitivity but a 65.5% specificity in identifying HCC. PSMA sensitivity remained high in the HCC tissue microarray (89.7%). PSMA was more accurate than CD34 (95.5% vs. 69.7%) in distinguishing grade 1 HCC from HA and high-grade DN while retaining high sensitivity (80%). The degree of PSMA positivity in HCC was greater in older, male, and human immunodeficiency virus patients ( P <0.05). No associations were found between PSMA staining and other tumor parameters ( P >0.05). PSMA is a marker of neoangiogenesis with increased expression in both primary and metastatic hepatic malignancies. Neovascular PSMA expression is more specific and accurate than CD34 for differentiating HCC from benign and precursor hepatic lesions. Diagnostic and therapeutic utility of PSMA radioligands in malignant liver neoplasms warrant further clinical investigations.
Collapse
Affiliation(s)
- Michel Kmeid
- Department of Pathology, Albany Medical Center, Albany
| | | | - Taek Chung
- Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Georgi Lukose
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Luz Sullivan
- Department of Pathology, Albany Medical Center, Albany
| | - Rupinder Brar
- Department of Pathology, Albany Medical Center, Albany
| | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany
| |
Collapse
|
5
|
Lennartz M, Gehrig E, Weidemann S, Gorbokon N, Menz A, Büscheck F, Hube-Magg C, Hinsch A, Reiswich V, Höflmayer D, Fraune C, Jacobsen F, Bernreuther C, Lebok P, Sauter G, Wilczak W, Steurer S, Burandt E, Marx AH, Simon R, Krech T, Clauditz TS, Minner S, Dum D, Uhlig R. Large-Scale Tissue Microarray Evaluation Corroborates High Specificity of High-Level Arginase-1 Immunostaining for Hepatocellular Carcinoma. Diagnostics (Basel) 2021; 11:diagnostics11122351. [PMID: 34943588 PMCID: PMC8699869 DOI: 10.3390/diagnostics11122351] [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: 11/01/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
Arginase-1 catalyzes the conversion of arginine to ornithine and urea. Because of its predominant expression in hepatocytes, it serves as a marker for hepatocellular carcinoma, although other tumor entities can also express arginase-1. To comprehensively determine arginase-1 expression in normal and neoplastic tissues, tissue microarrays containing 14,912 samples from 117 different tumor types and 608 samples of 76 different normal tissue types were analyzed by immunohistochemistry. In normal tissues, arginase-1 was expressed in the liver, the granular layer of the epidermis, and in granulocytes. Among tumors, a nuclear and cytoplasmic arginase-1 immunostaining was predominantly observed in hepatocellular carcinoma, where 96% of 49 cancers were at least moderately positive. Although 22 additional tumor categories showed occasional arginase immunostaining, strong staining was exceedingly rare in these entities. Staining of a few tumor cells was observed in squamous cell carcinomas of various sites. Staining typically involved maturing cells with the beginning of keratinization in these tumors and was significantly associated with a low grade in 635 squamous cell carcinomas of various sites (p = 0.003). Teratoma, urothelial carcinoma and pleomorphic adenomas sometimes also showed arginase expression in areas with squamous differentiation. In summary, arginase-1 immunohistochemistry is highly sensitive and specific for hepatocellular carcinoma if weak and focal staining is disregarded.
Collapse
Affiliation(s)
- Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Eva Gehrig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Andreas H. Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
- Department of Pathology, Academic Hospital Fuerth, 90766 Fuerth, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
- Correspondence: ; Tel.: +49-40-74105-7214
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
- Institute of Pathology, Clinical Center Osnabrueck, 49076 Osnabrueck, Germany
| | - Till S. Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.L.); (E.G.); (S.W.); (N.G.); (A.M.); (F.B.); (C.H.-M.); (A.H.); (V.R.); (D.H.); (C.F.); (F.J.); (C.B.); (P.L.); (G.S.); (W.W.); (S.S.); (E.B.); (A.H.M.); (T.K.); (T.S.C.); (S.M.); (D.D.); (R.U.)
| |
Collapse
|
6
|
Al-Shahari EA, El Barky AR, Mohamed TM, Alm-Eldeen AA. Doxorubicin, L-arginine, or their combination as a prophylactic agent against hepatic carcinoma in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:37661-37671. [PMID: 33721166 DOI: 10.1007/s11356-021-13177-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/22/2021] [Indexed: 06/12/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the ten most commonly diagnosed cancers. Doxorubicin is an antibiotic used in cancer treatment protocols that has several side effects. L-Arginine is a non-essential amino acid that is used as immune system activation and antitumor drugs. Therefore, the current study was designed to compare using doxorubicin, L-arginine, or their combination as a prophylactic agent against hepatic carcinoma induced by hepatocellular carcinoma cells (HepG2) injection in mice. The mice were divided into five groups: normal mice and mice that received HepG2, doxorubicin and HepG2, L-arginine and HepG2, and doxorubicin, L-Arginine, and HepG2, respectively. Liver function test as, aspartate transaminase (AST) and alanine transaminase (ALT), and alpha-fetoprotein (AFP), caspase 3, interleukin 6 (IL-6), tumor necrotic factor (TNF), lipid peroxidation (NDA), and some antioxidant parameters were determined. A significant increase in AST and ALT, α-fetoprotein, TNF-α, and cytokines IL6 and MDA and a significant decrease in the serum caspase and liver catalase were determined in HepG2-injected mice. Moreover, some large hyperchromatic heptocytes were observed and the percentage of the positive area/field of HepPar-1, the most specific HCC marker, was 9.56%. Interestingly, mice that received doxorubicin, L-arginine, or their combination showed an improvement in some of the previous parameters. The improvement was more prominent with L-arginine administration.
Collapse
Affiliation(s)
- Eman A Al-Shahari
- Department of Biology, Faculty of Science and Arts, King Khaled University, Abha, Saudi Arabia
- Department of Biology, Faculty of Science, Ibb University, Ibb, Yemen
| | - Amira Ragab El Barky
- Biochemistry Unit, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Tarek M Mohamed
- Biochemistry Unit, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | | |
Collapse
|
7
|
Patil PA, Taddei T, Jain D, Zhang X. HNF-1β is a More Sensitive and Specific Marker Than C-Reactive Protein for Identifying Biliary Differentiation in Primary Hepatic Carcinomas. Arch Pathol Lab Med 2021; 146:220-226. [PMID: 34086854 DOI: 10.5858/arpa.2020-0725-oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2021] [Indexed: 02/05/2023]
Abstract
CONTEXT.— Intrahepatic cholangiocarcinoma (iCCA) needs to be distinguished from hepatocellular carcinoma (HCC) and metastasis, and in the absence of any specific biliary markers, is often a diagnosis of exclusion. Hepatocyte nuclear factor (HNF)-1β is a transcription factor that plays a critical role in bile duct system morphogenesis. OBJECTIVE.— To investigate the diagnostic value of HNF-1β to differentiate iCCA from HCC by immunohistochemistry and compare HNF-1β with C-reactive protein (CRP), a previously identified marker for iCCA. DESIGN.— Cases of iCCA (n = 75), combined hepatocellular-cholangiocarcinoma (cHCC-CCA) (n = 13) and HCC (n = 65) were included in the study. RESULTS.— All cases of iCCA (74 of 74, 100%) expressed HNF-1β compared with CRP expressed in 72.60% (53 of 73). The sensitivity and specificity of HNF-1β to differentiate iCCA from HCC was 100% and 92.31%, whereas the sensitivity and specificity for CRP was 75.58% and 7.79%. The expression of HNF-1β was greater in iCCA and the CCA component of cHCC-CCA compared with CRP (87 of 87, 100% versus 65 of 86, 75.58%, P < .001). On the contrary, CRP was more frequently expressed compared with HNF-1β in HCC and HCC component of cHCC-CCA (71 of 77, 92.21% versus 6 of 78, 7.69%; P < .001). CONCLUSIONS.— Our data indicate that HNF-1β is a more sensitive and specific marker than CRP for the diagnosis of iCCA and to identify the CCA component in cHCC-CCA. Lack of HNF-1β expression may be used to exclude iCCA from consideration in cases of adenocarcinomas of unknown primary.
Collapse
Affiliation(s)
- Pallavi A Patil
- From the Department of Pathology (Patil, Jain, Zhang), Yale University School of Medicine, New Haven, Connecticut.,Patil is currently located in the Department of Pathology at the University of South Alabama, in Mobile, Alabama
| | - Tamar Taddei
- Section of Digestive Diseases (Taddei), Yale University School of Medicine, New Haven, Connecticut
| | - Dhanpat Jain
- From the Department of Pathology (Patil, Jain, Zhang), Yale University School of Medicine, New Haven, Connecticut
| | - Xuchen Zhang
- From the Department of Pathology (Patil, Jain, Zhang), Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
8
|
Arif D, Mettler T, Adeyi OA. Mimics of hepatocellular carcinoma: a review and an approach to avoiding histopathological diagnostic missteps. Hum Pathol 2020; 112:116-127. [PMID: 33346018 DOI: 10.1016/j.humpath.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/12/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the commonest carcinomas and leading causes of cancer-related death. Although, in patients with cirrhosis, radiologic diagnosis has improved significantly over the years, needle biopsy and histopathological assessment remains an important diagnostic modality. Most importantly, histopathological diagnosis is essential in patients with contending extrahepatic primaries, those with no known HCC risk factors, patients with ambiguous radiological features, and many other clinical contexts. Helpful features such as high serum alpha-fetoprotein (AFP) serologies are known to be present in many other tumor (including but not only HCC) and nontumor contexts and therefore not only lack sufficient diagnostic specificity for HCC but also create the potential to overlook non-HCC AFP-producing tumors, of which there are many. Therefore, using clinical examples and other examples from the medical literature, this review discusses several clinical and histological mimics of HCC and proffers an approach for practicing pathologists geared toward avoiding missteps.
Collapse
Affiliation(s)
- Dauod Arif
- University of Minnesota Medical School, Department of Laboratory Medicine & Pathology, MN 55455, USA
| | - Tetyana Mettler
- University of Minnesota Medical School, Department of Laboratory Medicine & Pathology, MN 55455, USA
| | - Oyedele A Adeyi
- University of Minnesota Medical School, Department of Laboratory Medicine & Pathology, MN 55455, USA.
| |
Collapse
|
9
|
Vyas M, Zhang X. Hepatocellular Carcinoma: Role of Pathology in the Era of Precision Medicine. Clin Liver Dis 2020; 24:591-610. [PMID: 33012447 DOI: 10.1016/j.cld.2020.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is a morphologically heterogeneous tumor with variable architectural growth patterns and several distinct histologic subtypes. Large-scale attempts have been made over the past decade to identify targetable genomic alterations in HCC; however, its translation into clinical personalized care remains a challenge to precision oncology. The role of pathology is no longer limited to confirmation of diagnosis when radiologic features are atypical. Pathology is now in a position to predict the underlying molecular alteration, prognosis, and behavior of HCC. This review outlines various aspects of histopathologic diagnosis and role of pathology in cutting-edge diagnostics of HCC.
Collapse
Affiliation(s)
- Monika Vyas
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 303 Brookline Avenue, Boston, MA 02215, USA
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, PO Box 208023, New Haven, CT 06520-8023, USA.
| |
Collapse
|
10
|
Chen DA, Koehne de Gonzalez A, Fazlollahi L, Coffey A, Remotti HE, Lagana SM. In situ hybridisation for albumin RNA in paediatric liver cancers compared with common immunohistochemical markers. J Clin Pathol 2020; 74:98-101. [PMID: 32471888 DOI: 10.1136/jclinpath-2020-206663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 11/04/2022]
Abstract
AIMS In situ hybridisation (ISH) for albumin mRNA is a sensitive marker of primary liver tumours in adults. However, paediatric tumours, such as hepatoblastoma (HB) and fibrolamellar hepatocellular carcinoma (FLC), have not been tested thoroughly and may require ancillary tests to diagnose with confidence. We aim to determine if albumin ISH is useful in the pathological evaluation of these malignancies and to compare it to commonly used immunohistochemical markers HepPar 1 (HEPA) and arginase-1 (ARG). METHODS Tissue microarrays of 26 HB and 10 FLC were constructed. Controls included 4 embryonal undifferentiated sarcomas of the liver, 51 neuroblastomas and 64 Wilms tumours. We evaluated a commercially available RNA ISH to detect albumin mRNA. Immunohistochemistry for HEPA and ARG was performed in the usual fashion. RESULTS Twenty-six of 26 HB showed positive staining by albumin ISH including 14 fetal, 8 embryonal and 4 mixed variants. All 10 FLC were diffusely positive. The sensitivity and specificity of albumin ISH were 100% for HB and FLC. ARG had 100% sensitivity and specificity for HB (26 of 26 cases) and FLC (9 of 9). HEPA stained 22 of 26 HB (85% sensitivity, 99.2% specificity) and 7 of 9 FLC (78% sensitivity, 99.1% specificity). CONCLUSION Albumin RNA ISH is a useful test to determine hepatocytic origin in HB and FLC. ARG was equally sensitive and easy to interpret, while HEPA was inferior to both in HB and FLC.
Collapse
Affiliation(s)
- Diane Ann Chen
- Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Anne Koehne de Gonzalez
- Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Ladan Fazlollahi
- Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Amy Coffey
- Department of Diagnostic Medicine, University of Texas System, Austin, Texas, USA
| | - Helen E Remotti
- Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Stephen M Lagana
- Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
11
|
Wang X, Xu Y, Wang R, Dai N, Zhang W, Li F. The significance of arginase-1 expression in the diagnosis of liver cancer: A protocol for a systematic review. Medicine (Baltimore) 2020; 99:e19159. [PMID: 32118719 PMCID: PMC7478447 DOI: 10.1097/md.0000000000019159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Pathologic distinction between HCC and intrahepatic cholangiocarcinoma (ICC) and metastatic adenocarcinoma can be challenging and sometimes requires immunohistochemical panels. Recently, arginase-1 (ARG-1) has been introduced for differentiation of these tumors. METHODS We will search Cochrane Library, PubMed, Embase, China National Knowledge Infrastructure through August 1, 2019, comprehensive collection studies about the diagnostic value of ARG-1 for HCC. Two reviewers will screen literature according to the inclusion and exclusion criteria, extract data, and assess the quality of included studies. Review Manager 5.3 and STATA 15.0 will be used to conduct the meta-analysis. RESULTS The review will provide a high-quality synthesis of current evidence of the diagnostic value of liver cancer. The results will be published in a peer-reviewed journal. CONCLUSION We hope that the results of this study will provide significant evidence to assess the value of ARG-1 in differential diagnosis of HCC, ICC, and metastatic carcinoma of liver.
Collapse
|
12
|
Obiorah IE, Chahine J, Park BU, Ko K, deGuzman J, Kallakury B. Well differentiated arginase-1 negative hepatocellular carcinoma. Transl Gastroenterol Hepatol 2019; 4:66. [PMID: 31620648 DOI: 10.21037/tgh.2019.08.01] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022] Open
Abstract
Background The diagnosis of hepatocellular carcinoma (HCC) is dependent on the histologic and immunohistochemical analysis of biopsy and resection specimens. The distinction of HCC from metastatic neoplasms is pertinent for treatment and prognostic purposes. Arginase-1 (Arg-1), a marker of hepatocellular differentiation, has shown superior sensitivity and specificity when compared to other immunohistochemical markers of detection of HCC such as hepatocyte paraffin antigen (HepPar-1). Studies have shown that poorly differentiated HCC can lose arginase expression, however well differentiated HCC are rarely ever arginase negative. Methods In this study Arg-1 expression was detected using immunohistochemical staining on tissue specimens from 40 confirmed cases of well differentiated HCC specimens using a highly specific monoclonal antibody for Arg-1. Specificity of the Arg-1 antibody was evaluated by immunostaining of 24 non-HCC tumors in the liver and 200 non-liver neoplasms using paraffin block and tissue micro-array (TMA) based immunohistochemistry. Results Four well differentiated HCC cases were found to be completely negative for Arg-1 and similarly all 224 non-HCC tumors did not express Arg-1. The arginase negative well differentiated tumors were positive for other hepatocellular markers such as HepPar-1 and polyclonal carcinoembryonic antigen (pCEA). Of the four tumors, only one recurred at 28 months. All patients are currently stable with a mean survival of 43 months. Conclusions Arg-1 negative well differentiated HCC can be a clinical dilemma which can lead to misdiagnosis. Confirmation with other hepatocellular markers such as HepPar1 and pCEA is essential in making the correct diagnosis. The clinicopathologic outcomes of arginase negative well differentiated HCC has been poorly characterized, thus our findings are of utmost importance in understanding the clinical behavior of these tumors. This may have a potential role in understanding the mechanism of the use of targeted therapy in HCC tumors.
Collapse
Affiliation(s)
- Ifeyinwa E Obiorah
- Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Joeffrey Chahine
- Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Byoung Uk Park
- Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Kyungmin Ko
- Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Jose deGuzman
- Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Bhaskar Kallakury
- Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, USA
| |
Collapse
|
13
|
Chen XB, Wei YH, Chen XK, Zhong J, Zou YB, Nie JY. Manganese levels and hepatocellular carcinoma: A systematic review and meta-analysis based on Asian cohort. Medicine (Baltimore) 2019; 98:e16748. [PMID: 31393389 PMCID: PMC6709027 DOI: 10.1097/md.0000000000016748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Several studies have investigated the relationship between Manganese (Mn) levels and hepatocellular carcinoma (HCC), but the results were inconsistent. Thus, we conducted a systematic review and meta-analysis to evaluate the association between Mn levels and HCC. Nine studies focusing on hair Mn levels, 6 studies on serum Mn levels and 6 studies on tissue Mn levels were identified in a systematic search of PubMed, CNKI, Wanfang and SinoMed databases. Standard mean differences (SMD) with the corresponding 95% confidence intervals (CI) were pooled to compare the Mn levels between HCC and controls. In serum, the Mn levels in HCC were significantly lower than in healthy controls (SMD (95% CI): -0.941 (-1.559, -0.323)). In hair, the Mn levels in HCC were slightly lower than in healthy controls, but not significant (SMD (95% CI): -0.168 (-0.766, 0.430)). In tissue, the Mn levels in tumors were significantly lower than in adjacent normal tissues (SMD (95% CI): -4.867 (-7.143, -2.592)). Subgroup analysis showed consistent results. In conclusion, this meta-analysis suggested an inverse association between Mn levels and HCC.
Collapse
Affiliation(s)
- Xiu-Bing Chen
- Department of Gastroenterology, The First People's Hospital of Qinzhou
| | - Yue-Hui Wei
- Quality Control Office, The Second People's Hospital of Qinzhou
| | - Xiu-Ke Chen
- Department of Cardiothoracic Surgery, The First People's Hospital of Qinzhou, Qinzhou, Guangxi
| | - Jian Zhong
- Department of Gastroenterology, The First People's Hospital of Qinzhou
| | - You-Bao Zou
- Department of Gastroenterology, The First People's Hospital of Qinzhou
| | - Jia-Yan Nie
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
14
|
Nasir A, Lehrke HD, Mounajjed T, Said S, Zhang L, Yasir S, Shah SS, Chandan VS, Smyrk TC, Moreira RK, Boland Froemming JM, Herrera Hernandez LP, Wu TT, Graham RP. Albumin In Situ Hybridization Can Be Positive in Adenocarcinomas and Other Tumors From Diverse Sites. Am J Clin Pathol 2019; 152:190-199. [PMID: 31107526 DOI: 10.1093/ajcp/aqz032] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Albumin messenger RNA (mRNA) expression is a marker of hepatocellular differentiation. Most published data are from review of tissue microarrays, and albumin in situ hybridization (ISH) expression across several tumor types is incompletely characterized. METHODS Sections from 221 tumors were evaluated for albumin mRNA. Immunohistochemistry was used to confirm diagnoses. Albumin ISH was performed according to manufacturer-provided instructions. Fifty-nine cases were evaluated with both commercial ISH assays. RESULTS Albumin mRNA was detected in all hepatocellular carcinomas (HCCs) and 81% of intrahepatic cholangiocarcinomas. Lung (20%), gallbladder (39%), hepatoid pancreatic (n = 1 of 1) adenocarcinoma, breast invasive ductal carcinoma (18%), yolk sac tumor (25%), and acinar cell carcinoma (29%) showed expression. Both assays were concordant in 93% of cases. CONCLUSIONS Albumin ISH was expressed in all HCCs studied. It was also positive in intrahepatic cholangiocarcinoma and patchy positive in gallbladder adenocarcinoma and a subset of other neoplasms, which can be a potential pitfall.
Collapse
Affiliation(s)
- Aqsa Nasir
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | - Heidi D Lehrke
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | | | - Samar Said
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | - Lizhi Zhang
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | - Saba Yasir
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | - Sejal S Shah
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | | | - Thomas C Smyrk
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | | | | | | | - Tsung-Teh Wu
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
15
|
Endogenous arginase 2 as a potential biomarker for PEGylated arginase 1 treatment in xenograft models of squamous cell lung carcinoma. Oncogenesis 2019; 8:18. [PMID: 30808864 PMCID: PMC6391460 DOI: 10.1038/s41389-019-0128-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Depletion of arginine induced by PEGylated arginase 1 (ARG1) (BCT-100) has shown anticancer effects in arginine auxotrophic cancers that lack argininosuccinate synthetase (ASS1) and ornithine transcarbamylase (OTC). High levels of endogenous arginase 2 (ARG2) have been previously reported in human lung cancers. Although a high-ARG2 level neither causes immunosuppression nor affects disease progression, it may theoretically affect the efficacy of PEGylated ARG1 treatment. ARG2 was shown to be highly expressed in H520 squamous cell lung carcinoma (lung SCC) xenografts but undetectable in SK-MES-1 and SW900 lung SCC xenografts. We propose that high-endogenous expression of ARG2 could impede the anti-tumor effect of PEGylated ARG1 in lung SCC. The in vivo effect of PEGylated ARG1 was investigated using three xenograft models of lung SCC. PEGylated ARG1 (60 mg/kg) suppressed tumor growth in SK-MES-1 and SW900 but not H520 xenografts. ASS1 was expressed in SK-MES-1 and SW900 xenografts while OTC expression remained low in all xenografts. A high-endogenous ARG2 level was detected only in H520 xenografts. Serum arginine level was decreased significantly by PEGylated ARG1 in all xenografts. Nonetheless intratumoral arginine level was decreased by PEGylated ARG1 in SK-MES-1 and SW900, not H520 xenografts. In SK-MES-1 xenografts, PEGylated ARG1 treatment induced G1 arrest, downregulation of Ki67 and Mcl-1 and activation of apoptosis. In SW900 xenografts, upregulation of Bim and activation of apoptosis were observed upon PEGylated ARG1 treatment. Silencing of ARG2 re-sensitized the H520 xenografts to PEGylated ARG1 treatment, partially mediated through arginine depletion via G1 arrest and apoptosis. PEGylated ARG1 treatment (BCT-100) was effective in lung SCC xenografts with low-endogenous levels of ASS1/OTC and ARG2. High-endogenous ARG2 expression may cause resistance to PEGylated ARG1 treatment in lung SCC xenografts. ARG2 may serve as a third predictive biomarker in PEGylated ARG1 treatment in lung SCC.
Collapse
|
16
|
Schlabritz-Loutsevitch N, Carrillo M, Li C, Nathanielsz P, Maguire C, Maher J, Dick E, Hubbard G, Stanek J. A first case of hepatocellular carcinoma in the baboon (Papio spp.) placenta. J Med Primatol 2018; 48:68-73. [PMID: 30246873 DOI: 10.1111/jmp.12382] [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: 05/18/2018] [Revised: 07/17/2018] [Accepted: 08/22/2018] [Indexed: 12/01/2022]
Abstract
We present a case of hepatocellular carcinoma (HCC) in the placenta of healthy baboon (Papio spp.). Grossly, the fetal, maternal, and placental tissues were unremarkable. Histologically, the placenta contained an unencapsulated, poorly demarcated, infiltrative, solidly cellular neoplasm composed of cells that resembled hepatocytes. The neoplastic cells were diffusely positive for vimentin and focally positive for Ae1/Ae3, Arginase -1, glutamine synthetase, and CD10, and negative for ER, vascular markers (CD31 and D240), S100, glypican, C-reactive protein, FABP, desmin, and beta-catenin; INI1 positivity was similar to non-neoplastic tissues. The case likely represents a unique subtype of HCC.
Collapse
Affiliation(s)
| | - Maira Carrillo
- Texas Tech University Health Sciences Center at the Permian Basin, Odessa, Texas
| | - Cun Li
- University of Wyoming, Laramie, Wyoming.,Texas Biomedical Research Institute, San Antonio, Texas
| | - Peter Nathanielsz
- University of Wyoming, Laramie, Wyoming.,Texas Biomedical Research Institute, San Antonio, Texas
| | - Christopher Maguire
- Texas Tech University Health Sciences Center at the Permian Basin, Odessa, Texas
| | - James Maher
- Texas Tech University Health Sciences Center at the Permian Basin, Odessa, Texas
| | - Edward Dick
- Texas Biomedical Research Institute, San Antonio, Texas
| | - Gene Hubbard
- University of Texas Health Sciences Center at San Antonio, San Antonio, Texas
| | | |
Collapse
|
17
|
Zhao CL, Hui Y, Wang LJ, Yang D, Yakirevich E, Mangray S, Huang CK, Lu S. Alanine-glyoxylate aminotransferase 1 (AGXT1) is a novel marker for hepatocellular carcinomas. Hum Pathol 2018; 80:76-81. [PMID: 29883780 DOI: 10.1016/j.humpath.2018.05.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/08/2018] [Accepted: 05/24/2018] [Indexed: 11/17/2022]
Abstract
Arginase-1 has been demonstrated as a marker for hepatocellular carcinoma (HCC) with higher sensitivity and specificity than HepPar-1 and glypican-3. However, its sensitivity is diminished in moderately and poorly differentiated HCCs. In the current study, we evaluated the utility of AGXT1 as a diagnostic marker. Immunostains for AGXT1 and arginase-1 were performed in tissue microarrays of 139 HCCs and 374 gastrointestinal and nongastrointestinal carcinomas. AGXT1 exhibited granular cytoplasmic immunoreactivity in contrast to the diffuse cytoplasmic staining characteristic of arginase-1 in nonneoplastic and neoplastic hepatocytes. Sensitivities of AGXT1 for all HCCs were 90.0% compared to 87.8% for arginase-1. A small number of tumors expressed only 1 of the 2 markers. Sensitivity increased to 92.1% when the presence of either marker was considered positive. Excepting 5 cases of cholangiocarcinoma, both AGXT1 and arginase-1 were negative in all non-HCC tumors with specificities of 98.7%. Our data support the consideration of AGXT1 as a novel hepatocellular marker with equally high specificity and slightly higher sensitivity as compared to arginase-1. AGXT1 may aid in diagnostic workup especially in conjunction with arginase-1 for HCCs that may otherwise defy conventional immunostaining patterns.
Collapse
Affiliation(s)
- Chaohui Lisa Zhao
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903
| | - Yiang Hui
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903
| | - Li Juan Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903
| | - Evgeny Yakirevich
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903
| | - Shamlal Mangray
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903
| | - Chiung-Kuei Huang
- Liver Research Center, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903.
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903.
| |
Collapse
|