RSS-Feed abonnieren

DOI: 10.1055/s-0044-1789589
Role of Dysregulated Immune Biomarkers in Hepatocellular Carcinoma Metastasis: A Systematic Meta-Analysis Review
- Introduction
- Methodology
- Data Extraction
- Statistical Analysis
- Results
- Quantitative Synthesis (Meta-Analysis)
- Discussion
- Conclusion
- References
Abstract


The liver plays a crucial role in immune system regulation, but dysregulation of immunological networks contributes to chronic liver diseases like hepatocellular carcinoma. This malignant tumor is the third leading cause of cancer death. An imbalanced immune system, characterized by alterations in immune cell count, cytokine levels, and inhibitory receptors, can impact metastasis by suppressing the immune system's ability to fight cancer cells. This study aims to investigate the potential biomarkers playing a crucial role in immune dysregulation resulting in hepatocellular carcinoma metastasis. A comprehensive and systematic literature review was conducted using both free words and search terms. The data extraction was then performed by a thorough literature screening. Next, the meta-analysis was performed using the metabin function of the meta library in R to evaluate the patient cases reporting metastasis in the event group. A total of 1,008 cases were considered, with 357 as events and 651 as nonevents. The results of the meta-analysis demonstrated the significant role of biomarkers in immune dysregulation causing metastasis (risk ratio = 0.54, 95% confidence interval: 0.4972, 0.6048, I 2 = 92.4%, p < 0.01). In addition to the immune dysregulation explored in this study, the impact of tumor size on hepatocellular carcinoma progression and metastasis is a crucial consideration. A notable difference of 41 more cases was reported for larger tumor sizes. The study integrates immune dysregulation biomarkers and tumor size factors influencing hepatocellular carcinoma metastasis, offering valuable insights for future research and therapeutic interventions for improved clinical outcomes.
Keywords
metastasis - hepatocellular carcinoma - dysregulated gene - immune response to cancer - cancer biomarkerIntroduction
The liver plays a crucial role in modulating the immune system, ensuring its protection via immunotolerance.[1] The liver plays a significant role in eliminating microbial compounds and pathogens from the bloodstream, along with detecting and removing infectious organisms.[2] The dysregulation of immunological networks is a crucial factor in the development and progression of chronic liver diseases and hepatocellular carcinoma (HCC).[3] [4] [5]
HCC is considered as one of the most prevalent malignant tumors and the third leading cause of cancer death.[6] [7] Hepatocellular carcinogenesis involves numerous genetic and epigenetic changes that are influenced by environmental factors. C-myc, cyclin D1, p53, p16, E-cadherin, and PTEN genes have been linked to hepatocarcinogenesis.[8] The experimental research data indicated a correlation between HCC development and exposure to hepatotropic virus infection, aflatoxin, algal hepatotoxins in contaminated water, and alcohol abuse resulting in liver cirrhosis.[9] The primary underlying cause of HCC is infection with the hepatitis B virus, especially in individuals with the aforementioned risk factors.[10] Historically, patients with HCC have had a poor prognosis primarily due to late diagnosis and high rates of recurrence. The treatment options currently available have limited success rates, with approximately 50% of patients experiencing a recurrence within 3 years after surgery. The long-term survival rate is also quite low, ranging from 30 to 40% at 5 years postsurgery.[11]
Various factors contribute to the development of tumor antigen tolerance, including reduced recognition of cancerous cells, immune suppression, and chronic inflammation caused by either a viral infection or immune dysregulation, ultimately contributing to the formation of cancer.[12] [13] Recent studies have shown that an imbalanced immune system, characterized by alterations in immune cell count or activity, cytokine levels, and the presence of inhibitory receptors or their ligands, plays a significant role in the progression of HCC.[14] The immune response can be shifted toward tumor tolerance due to changes in the function or expression of immune components, leading to the progression of the tumor. Various immune cells, including cytotoxic T cells, CD4+ T cells, regulatory T cells (Tregs), myeloid-derived suppressor cells, natural killer cells, and their interactions, have been found to play a role in the progression of HCC.[15] The interactions are sustained by proinflammatory cytokines and anti-inflammatory cytokines.[10]
Interleukins (ILs), also known as cytokines, regulate inflammatory and immune responses by activating and regulating immune cells and participating in an inflammatory cascade. Moreover, chemokines, the largest member of the cytokine family, may serve pivotal roles in inducing organ-specific metastasis.[16] [17] [18] After the establishment of HCC, various cytokines are released by the tumor itself, nearby nontumor cells, or immune cells, acting on the malignant lesion and supporting tumor survival through multiple mechanisms.[19] [20]
The impact of chemokines and their receptors on HCC varies, encompassing both the progression and inhibition of tumor growth. Cytokines control the recruitment of white blood cells and have a vital function in various processes, including angiogenesis, Th1/Th2 development, inflammatory diseases, and tumor growth, having a strong association with HCC and correlating with distant organs and lymph nodes metastasis.[1] The dysregulation of proinflammatory cytokines in the tumor microenvironment can impact metastasis by suppressing the immune system's ability to fight against cancer cells and inhibiting the activity of metastasis suppressors.[21] Furthermore, these cytokines have the ability to influence the tumor microenvironment, leading to immune evasion and metastasis,[22] making it necessary to identify new targets for the early detection and treatment of the disease.[23] [24]
This systematic review and meta-analysis aim to comprehensively evaluate existing research studies focusing on intricate connections between immune dysregulation, biomarkers, and cytokine signaling in HCC metastasis, offering valuable insights for advancing clinical strategies and improving outcomes in patients. A deeper understanding of the dysregulation of proinflammatory cytokines and their impact on immune evasion and metastasis may help identify new therapeutic strategies.
Methodology
Literature Search
An extensive literature review was conducted in databases such as PubMed and Google Scholar for articles published in the last 20 years to investigate the role of immune dysregulation and biomarkers involved in HCC metastasis. The literature review comprised both subject terms and free words. The free words used for the search were: “HCC,” “hepatocellular carcinoma,” “immune dysregulation,” and “metastasis,” along with the (“HCC” AND “metastasis” AND “cytokines”), (“HCC” AND “metastasis” AND “interferon”), (“HCC” AND “metastasis” AND “TLR”), (“HCC” AND “metastasis” AND “TNF”), and (“HCC” AND “metastasis” AND “antigens”) subject terms.
Inclusion Criteria
(1) Studies reporting HCC metastasis occurrence in patients.
(2) Studies considering any type of immune dysregulation of genes involved in HCC.
(3) Studies reporting biomarkers causing metastasis in HCC.
(4) Experimental and retrospective studies were primarily considered.
Exclusion Criteria
(1) Studies that did not clearly report HCC metastasis.
(2) Studies without open access were excluded.
(3) Studies without full-text availability were excluded.
Data Extraction
Literature screening was performed based on predefined inclusion and exclusion criteria. Data from each study investigating the HCC metastasis was extracted. Notably, the data extracted from each study included first author name and publication year, patient characteristics, sample sizes (n), event (patients reporting occurrence of metastasis), and nonevent (patients that did not report occurrence of metastasis). Moreover, the tumor size was also considered to evaluate the metastasis risk. Each dysregulated gene was further categorized into their respective families. The data also included findings of each gene to understand its potential in immune dysregulation resulting in HCC metastasis. This study aimed to gather a list of biomarkers responsible for immune dysregulation, further leading to HCC metastasis. A comprehensive range of evidence was considered in this meta-analysis.
Statistical Analysis
The “metabin” function of the “meta” library in R was used to conduct the meta-analysis. The function requires the number of patients reporting metastasis as the event group, the patients that did not report any metastasis as the nonevent group, and the sample sizes of the groups. Statistical analysis was conducted to obtain insights into the combined study outcomes. This involved calculating effect sizes, confidence intervals (CIs), weight percentages, and measures of heterogeneity (such as I 2 and τ 2). A statistical significance assessment was performed with a significance threshold set at p < 0.05. Moreover, the publication bias was assessed using Egger's test.
Results
Study Characteristics
Four sequential steps were followed for literature screening: identification, screening, eligibility, and final inclusion, illustrated in [Fig. 1]. After a thorough screening of 64 papers and the removal of duplicate studies, a total of 10 which met the criteria were selected. The study aimed to investigate the biomarkers that lead to immune dysregulation, eventually resulting in HCC metastasis. Further analyses were performed on these studies.


The final studies ranged from 2009 onwards. This meta-analysis included a total of 1,008 patients (850 males and 158 females), comprising 357 patients in the event group and 651 patients in the nonevent group. Moreover, each study classified patients based on an average age of 58 years, creating two distinct groups: one comprising individuals with an average age greater than 58 years and the other with an average age less than 58 years. However, the studies conducted by Yang et al and Zhang et al lacked a clear division of patients into age groups, reporting an average age of 52 and 56 years for both groups, respectively. The clinical data of the patients is represented in [Table 1]. Furthermore, the biomarkers were considered in each study. These genes were then divided into their respective family categories. Notably, six genes were from the chemokine family, two were from the cytokines family, and the remaining two were from the deubiquitinating enzyme (DUB) and antigen family, respectively. The baseline patient characteristics, such as age and gender, are depicted in [Fig. 2].


Study Id |
Sample size (n) |
Sex |
Age (y) |
References |
|||
---|---|---|---|---|---|---|---|
Event |
Nonevent |
Male |
Female |
≤ 58 |
> 58 |
||
Li et al (2012)[25] |
28 |
53 |
64 |
17 |
46 |
35 |
(25) |
Li et al (2015)[26] |
32 |
70 |
87 |
15 |
49 |
53 |
(26) |
Xue et al (2013)[27] |
24 |
38 |
56 |
6 |
47 |
15 |
(27) |
Li et al (2017)[28] |
40 |
38 |
67 |
11 |
54 |
24 |
(28) |
Lan et al (2014)[29] |
22 |
23 |
31 |
14 |
30 |
15 |
(29) |
Yang et al (2012)[30] |
69 |
219 |
254 |
34 |
52 |
(30) |
|
Li et al (2023)[31] |
15 |
70 |
70 |
15 |
54 |
31 |
(31) |
Kang et al (2018)[32] |
20 |
25 |
39 |
6 |
20 |
25 |
(32) |
Gao et al (2020)[33] |
52 |
28 |
63 |
17 |
35 |
45 |
(33) |
Zhang et al (2009)[34] |
55 |
87 |
119 |
23 |
56 |
(34) |
Immune Biomarkers across Studies
A comprehensive literature search was performed to enlist the biomarkers responsible for immune dysregulation causing HCC metastasis. This meta-analysis encompasses a total of 10 genes from various families, including two cytokine genes (IL23 and IL17A), six chemokine genes (CXCL8, CXCR7, CXCL13, CXCL9, CCL22, and IP10), one DUB gene (USP13), and one antigen gene (MAGE1). The distribution ratio of these genes into their respective families across the studies is listed in [Table 2] and illustrated in [Fig. 3].


Tumor Size across Studies
The distribution of tumor sizes categorized as 5 cm or less and greater than 5 cm was reported across studies. In this meta-analysis, a total of 617 cases of tumor size data was extracted, where 330 were reported to be less or equal to 5 cm and 287 greater than 5 cm. Four studies by Yunzhi Dang, Li et al, Kang et al, and Zhang et al reported cases with tumor sizes less than 5 cm.[57] Conversely, five studies by Li et al, Xue et al, Li et al, Lan et al, and Gao et al reported higher cases of tumor sizes greater than 5 cm. Notably, the study conducted by Yang et al did not provide any specific information regarding tumor sizes. A comprehensive overview of the studies, accompanied by the corresponding patient cases and reported tumor sizes, is represented in [Table 3]. A quantitative analysis was conducted to compare the reported tumor sizes among patient cases. The study by Zhang et al was excluded from the analysis due to its aberrant data, leading to a significantly higher number of cases with tumor sizes greater than 5 cm. The comparative analysis of the remaining studies is illustrated in [Fig. 4].


Abbreviation: NA, not available.
Quantitative Synthesis (Meta-Analysis)
Role of Dysregulated Immune Biomarkers in Hepatocellular Carcinoma Metastasis
All 10 studies reported the biomarkers responsible for immune dysregulation causing the occurrence of HCC metastasis in the event group. The data set representing 357 number of events from 1,008 total samples, along with 651 nonevents and their respective outcome ratios, are represented in [Table 4]. The patient cases reporting metastasis were considered as events within the total sample size. Only studies by Li et al and Gao et al reported a higher number of events while the remaining reported higher nonevents. The meta-analysis results showed significant heterogeneity in the role of biomarkers in immune dysregulation causing HCC metastasis (risk ratio = 0.54, 95% CI: 0.4972, 0.6048, I 2 = 92.4%, p < 0.01). The publication bias analysis reported insignificant results (test result: t = 0.90, degrees of freedom = 8, p-value = 0.39). The meta-analysis results are depicted in [Fig. 5].


Discussion
HCC is a common lethal cancer globally, particularly in areas with a high prevalence of chronic viral hepatitis infection, notably hepatitis B infection, causing both intrahepatic and extrahepatic metastases. HCC often metastases to the lungs, lymph nodes, adrenal gland, and bones, including the skull.[35] [36] Factors such as immune dysregulation lead to carcinogenesis.[13] The functions of diverse immune cells undergo dysregulation as the disease progresses from liver cirrhosis to HCC. This dysregulation manifests as a decreased secretion of Th1 cytokines, leading to a reduction in the antigen presentation capabilities of dendritic cells, playing a crucial role in the development of HCC.[15] Alterations in immune function or expression can cause the immune response to shifting toward tumor tolerance, leading to the progression of the tumor.[14] [37]
Chemokines, a family of small cytokines, are crucial in coordinating immune cell trafficking and shaping the immune profile of tumors. They influence the migration patterns of immune cells into tumors, often favoring a protumorigenic state, directly impacting the nonimmune cells within the tumor microenvironment, including cancer cells, stromal cells, and vascular endothelial cells. Given their integral role in the tumor immune response and tumor biology, the chemokine network has emerged as a potential target for immunotherapy. Hence, in this study, 10 genes, particularly those encoding cytokines (IL23 and IL17A) and chemokines (CXCL8, CXCR7, CXCL13, CXCL9, and CCL22), as well as DUB (USP13) and antigen (MAGE1), have been identified as implicated in dysregulation, influencing immune cell behavior within the tumor microenvironment.
The aforementioned genes are intimately linked to both immune dysregulation and the metastatic progression of HCC. Notably, the dysregulation of IL23 and IL17A cytokines has been observed to actively promote tumorigenesis.[38] [39] Additionally, in a murine HCC model generated using a specific HCC cell line, the overexpression of IL23 resulted in enhanced growth of HCC, demonstrating a dependence on IL17 for this effect,[40] underscoring the intricate relationship between these cytokines and their consequential impact on HCC progression, as evident in the experimental mouse model.[41]
Moreover, the chemokine family genes, including CXCL8, CXCR7, CXCL13, CXCL9, and CCL22, activate the signaling pathways for the development of tumor diseases.[42] The CXCL family not only regulates leukocyte migration but also controls tumor growth, immune cell activity, tumor cell proliferation, invasion, neoplastic microvascular formation, and tumor cell transformation, altering the angiogenic environment and promoting local tumor cell growth and metastasizes to distant organs. The role of the CXCL family is closely related to HCC.[43] The upregulation of CXCL8 and CXCL9 promoted tumor cell proliferation, migration, and metastasis through the interaction between HCC cells and macrophages. Moreover, these genes are also considered potential biomarkers of chronic hepatitis B.[44] [45]
The CXCL13, identified as a B-cell chemoattractant, plays fundamental roles in inflammatory, infectious, and immune responses.[46] While the CXCL13 gene has been previously associated with prostate cancer cell proliferation and abnormal signaling activation, recent findings suggest its potential involvement in the development and progression of HCC due to higher serum levels.[28] [47] [48] Furthermore, the study by Zheng et al reported that inhibition of CXCR7, a chemokine receptor, decreases angiogenesis and tumor growth in the murine HCC model.[49] It is noteworthy that CCL22 plays a role in recruiting Treg cells, and heightened levels of this chemokine correlate with poorer survival outcomes in individuals with HCC.[50]
Lastly, the USP13, a deubiquitination enzyme, is involved in various cellular processes like mitochondrial energy metabolism, autophagy, deoxyribonucleic acid damage response, and endoplasmic reticulum-associated degradation,[51] whereas MAGE1, with strong tumor antigen properties, holds significance in immune responses against cancer cells. Thus, the dysregulation of USP13 and MAGE1 is reported in many malignant tumors, including HCC.[52] [53] The reported dysregulation of USP13 and MAGE1, encompassing their involvement in diverse cellular processes and immune responses, underlines their potential significance in the metastasis of HCC. Hence, in this meta-analysis, 10 studies consisting of the aforementioned genes played a crucial role as biomarkers in immune dysregulation within the tumor microenvironment, causing HCC metastasis. All studies, except Li et al and Gao et al, reported higher cases of metastasis from the sample size. A significant heterogeneity was observed among studies with insignificant publication bias.
Additionally, a quantitative analysis of tumor parameter (tumor size 5 and > 5 cm) data was also extracted across studies. Tumor size is an independent predictor of HCC progression, metastases formation, and overall survival. Tumor size exhibits a direct correlation with the likelihood of metastasis. As the size of the tumor increases, the probability of metastatic spread also increases.[54] [55] [56] Four studies reported a higher incidence of cases for tumors measuring 5 cm or less, whereas five studies reported larger tumor sizes, indicating a difference of 41 more cases in the category of larger tumor sizes. Only one study by Yang et al did not provide information on tumor size. Despite some studies focusing on tumor sizes of 5 cm or less, it is noteworthy that all studies across both categories consistently reported cases of metastasis.
This pooled analysis of biomarkers in immune dysregulation within the tumor microenvironment has highlighted the intricate interplay between various cytokines, chemokines, and regulatory elements, emphasizing their crucial roles in HCC metastasis. Additionally, the quantitative analysis of tumor size reaffirms it as an independent predictor of HCC progression and metastasis. The correlation between larger tumor sizes and an increased likelihood of metastatic spread, as evidenced by the observed difference in cases across studies, highlights the clinical relevance of tumor size as a prognostic factor.
However, it is important to acknowledge certain limitations within the meta-analysis. A notable constraint lies in the variability among studies regarding the clear division of patient cases into HCC metastasis events or nonevents. Furthermore, the absence of a comprehensive understanding of the specific pathways and interactions through which these dysregulated genes directly contribute to metastasis underscores a current limitation in the studies. Future studies with a more focused exploration of the functional implications of these genes in the metastatic process will be crucial for unraveling the underlying biology and advancing targeted therapeutic strategies for HCC. Despite these limitations, this meta-analysis provides valuable insights into the complex relationship between biomarkers, tumor size, and HCC metastasis, emphasizing the need for standardized reporting practices and comprehensive data in future studies.
Conclusion
In conclusion, the comprehensive meta-analysis of the 10 selected studies sheds light on the intricate relationship underlying immune dysregulation and metastatic progression in HCC. The identified genes, encompassing cytokines (IL23 and IL17A), chemokines (CXCL8, CXCR7, CXCL13, CXCL9, and CCL22), as well as DUB (USP13) and antigen (MAGE1), have emerged as crucial players in influencing immune cell behavior within the tumor microenvironment. These biomarkers exhibit significant associations with both immune dysregulation and HCC metastasis. The heterogeneity observed among the studies, along with the absence of publication bias, underscores the robustness and reliability of the meta-analysis results. Moreover, the quantitative analysis of tumor parameters, specifically tumor size, reaffirms its role as an independent predictor of HCC progression and metastasis. Although this meta-analysis findings underscore the importance of these biomarkers as potential targets for therapeutic interventions, the limitations should be considered for comprehensive evaluation and validation of the results.
Conflict of Interest
None declared.
Availability of Data and Materials
All data throughout the manuscript are available in manuscript body and [Supplementary Material] (available in online version only).
-
References
- 1 Taibi C, Vincenzi L, D'Offizi G. Role of the immune system in hepatocellular carcinoma. In: Ettorre GM. ed. Hepatocellular Carcinoma [Internet]. Cham:: Springer International Publishing;; 2023: 19-26
- 2 Øie CI, Wolfson DL, Yasunori T. et al. Liver sinusoidal endothelial cells contribute to the uptake and degradation of entero bacterial viruses. Sci Rep 2020; 10 (01) 898
- 3 Ringelhan M, Pfister D, O'Connor T, Pikarsky E, Heikenwalder M. The immunology of hepatocellular carcinoma. Nat Immunol 2018; 19 (03) 222-232
- 4 Cho HJ, Cheong JY. Role of immune cells in patients with hepatitis B virus-related hepatocellular carcinoma. Int J Mol Sci 2021; 22 (15) 8011
- 5 Yu LX, Ling Y, Wang HY. Role of nonresolving inflammation in hepatocellular carcinoma development and progression. NPJ Precis Oncol 2018; 2 (01) 6
- 6 Hatziapostolou M, Polytarchou C, Aggelidou E. et al. An HNF4α-miRNA inflammatory feedback circuit regulates hepatocellular oncogenesis. Cell 2011; 147 (06) 1233-1247
- 7 Mínguez B, Hoshida Y, Villanueva A. et al. Gene-expression signature of vascular invasion in hepatocellular carcinoma. J Hepatol 2011; 55 (06) 1325-1331
- 8 Villanueva A, Newell P, Chiang DY, Friedman SL, Llovet JM. Genomics and signaling pathways in hepatocellular carcinoma. Semin Liver Dis 2007; 27 (01) 55-76
- 9 Poon D, Anderson BO, Chen LT. et al; Asian Oncology Summit. Management of hepatocellular carcinoma in Asia: consensus statement from the Asian Oncology Summit 2009. Lancet Oncol 2009; 10 (11) 1111-1118
- 10 Papatheodoridis GV, Lampertico P, Manolakopoulos S, Lok A. Incidence of hepatocellular carcinoma in chronic hepatitis B patients receiving nucleos(t)ide therapy: a systematic review. J Hepatol 2010; 53 (02) 348-356
- 11 Aravalli RN, Steer CJ, Cressman ENK. Molecular mechanisms of hepatocellular carcinoma. Hepatology 2008; 48 (06) 2047-2063
- 12 Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010; 140 (06) 883-899
- 13 Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011; 331 (6024) 1565-1570
- 14 Aravalli RN. Role of innate immunity in the development of hepatocellular carcinoma. World J Gastroenterol 2013; 19 (43) 7500-7514
- 15 Sachdeva M, Chawla YK, Arora SK. Immunology of hepatocellular carcinoma. World J Hepatol 2015; 7 (17) 2080-2090
- 16 Xue D, Zheng Y, Wen J. et al. Role of chemokines in hepatocellular carcinoma (Review). Oncol Rep 2021; 45 (03) 809-823
- 17 Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol 2017; 17 (09) 559-572
- 18 Kakinuma T, Hwang ST. Chemokines, chemokine receptors, and cancer metastasis. J Leukoc Biol 2006; 79 (04) 639-651
- 19 Makarova-Rusher OV, Medina-Echeverz J, Duffy AG, Greten TF. The yin and yang of evasion and immune activation in HCC. J Hepatol 2015; 62 (06) 1420-1429
- 20 Fu Y, Liu S, Zeng S, Shen H. From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma. J Exp Clin Cancer Res 2019; 38 (01) 396
- 21 Karin M. Nuclear factor-kappaB in cancer development and progression. Nature 2006; 441 (7092) 431-436
- 22 Cabillic F, Corlu A. Regulation of transdifferentiation and retrodifferentiation by inflammatory cytokines in hepatocellular carcinoma. Gastroenterology 2016; 151 (04) 607-615
- 23 Frau M, Biasi F, Feo F, Pascale RM. Prognostic markers and putative therapeutic targets for hepatocellular carcinoma. Mol Aspects Med 2010; 31 (02) 179-193
- 24 Silva MF, Sherman M. Criteria for liver transplantation for HCC: what should the limits be?. J Hepatol 2011; 55 (05) 1137-1147
- 25 Li J, Lau G, Chen L. et al. Interleukin 23 promotes hepatocellular carcinoma metastasis via NF-kappa B induced matrix metalloproteinase 9 expression. PLoS One 2012; 7 (09) e46264
- 26 Li XP, Yang XY, Biskup E. et al. Co-expression of CXCL8 and HIF-1α is associated with metastasis and poor prognosis in hepatocellular carcinoma. Oncotarget 2015; 6 (26) 22880-22889
- 27 Xue TC, Chen RX, Ren ZG, Zou JH, Tang ZY, Ye SL. Transmembrane receptor CXCR7 increases the risk of extrahepatic metastasis of relatively well-differentiated hepatocellular carcinoma through upregulation of osteopontin. Oncol Rep 2013; 30 (01) 105-110
- 28 Li B, Su H, Cao J, Zhang L. CXCL13 rather than IL-31 is a potential indicator in patients with hepatocellular carcinoma. Cytokine 2017; 89: 91-97
- 29 Lan X, Xiao F, Ding Q. et al. The effect of CXCL9 on the invasion ability of hepatocellular carcinoma through up-regulation of PREX2. J Mol Histol 2014; 45 (06) 689-696
- 30 Yang P, Li QJ, Feng Y. et al. TGF-β-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell 2012; 22 (03) 291-303
- 31 Li Y, Wang C, Yin X, Jiang L, Li X, Yang J. Profile and clinical significance of interferon gamma-inducible protein-10 (IP-10) and its receptor in patients with hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149 (16) 14879-14888
- 32 Kang Y, Su G, Sun J, Zhang Y. Activation of the TLR4/MyD88 signaling pathway contributes to the development of human hepatocellular carcinoma via upregulation of IL-23 and IL-17A. Oncol Lett 2018; 15 (06) 9647-9654
- 33 Gao S, Chen T, Li L. et al. Hypoxia-inducible ubiquitin specific peptidase 13 contributes to tumor growth and metastasis via enhancing the Toll-like receptor 4/myeloid differentiation primary response gene 88/nuclear factor-κB pathway in hepatocellular carcinoma. Front Cell Dev Biol 2020; 8: 587389
- 34 Zhang Y, Song T, Meng L, Wu X, Ba Y, Li Q. Melanoma antigen-1 mRNA combined with α-fetoprotein mRNA levels in peripheral blood of patients with hepatocellular carcinoma: a predictor of postoperative recurrence or metastasis?. ANZ J Surg 2009; 79 (1-2): 62-69
- 35 Targe M, Yasam VR, Nagarkar R. Hepatocellular carcinoma with uncommon sites of metastasis: a rare case report. Egypt J Radiol Nucl Med 2021; 52 (01) 228
- 36 Kummar S, Shafi NQ. Metastatic hepatocellular carcinoma. Clin Oncol (R Coll Radiol) 2003; 15 (05) 288-294
- 37 Matsui M, Machida S, Itani-Yohda T, Akatsuka T. Downregulation of the proteasome subunits, transporter, and antigen presentation in hepatocellular carcinoma, and their restoration by interferon-γ. J Gastroenterol Hepatol 2002; 17 (08) 897-907
- 38 Chan IH, Jain R, Tessmer MS. et al. Interleukin-23 is sufficient to induce rapid de novo gut tumorigenesis, independent of carcinogens, through activation of innate lymphoid cells. Mucosal Immunol 2014; 7 (04) 842-856
- 39 Kirchberger S, Royston DJ, Boulard O. et al. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J Exp Med 2013; 210 (05) 917-931
- 40 Liu Y, Song Y, Lin D. et al. NCR- group 3 innate lymphoid cells orchestrate IL-23/IL-17 axis to promote hepatocellular carcinoma development. EBioMedicine 2019; 41: 333-344
- 41 Warner K, Ghaedi M, Chung DC, Jacquelot N, Ohashi PS. Innate lymphoid cells in early tumor development. Front Immunol 2022; 13: 948358
- 42 Wu T, Yang W, Sun A, Wei Z, Lin Q. The role of CXC chemokines in cancer progression. Cancers (Basel) 2022; 15 (01) 167
- 43 Zajkowska M, Mroczko B. Chemokines in primary liver cancer. Int J Mol Sci 2022; 23 (16) 8846
- 44 Yin Z, Huang J, Ma T. et al. Macrophages activating chemokine (C-X-C motif) ligand 8/miR-17 cluster modulate hepatocellular carcinoma cell growth and metastasis. Am J Transl Res 2017; 9 (05) 2403-2411
- 45 Yu X, Chen Y, Cui L. et al. CXCL8, CXCL9, CXCL10, and CXCL11 as biomarkers of liver injury caused by chronic hepatitis B. Front Microbiol 2022; 13: 1052917
- 46 Kazanietz MG, Durando M, Cooke M. CXCL13 and its receptor CXCR5 in cancer: inflammation, immune response, and beyond. Front Endocrinol (Lausanne) 2019; 10: 471
- 47 Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm 2022; 3 (02) e147
- 48 Zhou C, Gao Y, Ding P, Wu T, Ji G. The role of CXCL family members in different diseases. Cell Death Discov 2023; 9 (01) 212
- 49 Zheng K, Li HY, Su XL. et al. Chemokine receptor CXCR7 regulates the invasion, angiogenesis and tumor growth of human hepatocellular carcinoma cells. J Exp Clin Cancer Res 2010; 29 (01) 31
- 50 Zhu F, Li X, Chen S, Zeng Q, Zhao Y, Luo F. Tumor-associated macrophage or chemokine ligand CCL17 positively regulates the tumorigenesis of hepatocellular carcinoma. Med Oncol 2016; 33 (02) 17
- 51 Li X, Yang G, Zhang W. et al. USP13: multiple functions and target inhibition. Front Cell Dev Biol 2022; 10: 875124
- 52 Huang J, Gu ZL, Chen W, Xu YY, Chen M. Knockdown of ubiquitin-specific peptidase 13 inhibits cell growth of hepatocellular carcinoma by reducing c-Myc expression. Kaohsiung J Med Sci 2020; 36 (08) 615-621
- 53 Li R, Gong J, Xiao C. et al. A comprehensive analysis of the MAGE family as prognostic and diagnostic markers for hepatocellular carcinoma. Genomics 2020; 112 (06) 5101-5114
- 54 Liu C, Xiao GQ, Yan LN. et al. Value of α-fetoprotein in association with clinicopathological features of hepatocellular carcinoma. World J Gastroenterol 2013; 19 (11) 1811-1819
- 55 Yoo DJ, Kim KM, Jin YJ. et al. Clinical outcome of 251 patients with extrahepatic metastasis at initial diagnosis of hepatocellular carcinoma: does transarterial chemoembolization improve survival in these patients?. J Gastroenterol Hepatol 2011; 26 (01) 145-154
- 56 Jun L, Zhenlin Y, Renyan G. et al. Independent factors and predictive score for extrahepatic metastasis of hepatocellular carcinoma following curative hepatectomy. Oncologist 2012; 17 (07) 963-969
- 57 Dang Y, Chen J, Feng W, Qiao C. et al. Interleukin 1β-mediated HOXC10 overexpression promotes hepatocellular carcinoma metastasis by upregulating PDPK1 and VASP. Theranostics 2020; 10 (08) 3833
Address for correspondence
Publikationsverlauf
Eingereicht: 15. Mai 2024
Angenommen: 26. Juli 2024
Artikel online veröffentlicht:
26. August 2024
© 2024. MedIntel Services Pvt Ltd. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)
Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India
-
References
- 1 Taibi C, Vincenzi L, D'Offizi G. Role of the immune system in hepatocellular carcinoma. In: Ettorre GM. ed. Hepatocellular Carcinoma [Internet]. Cham:: Springer International Publishing;; 2023: 19-26
- 2 Øie CI, Wolfson DL, Yasunori T. et al. Liver sinusoidal endothelial cells contribute to the uptake and degradation of entero bacterial viruses. Sci Rep 2020; 10 (01) 898
- 3 Ringelhan M, Pfister D, O'Connor T, Pikarsky E, Heikenwalder M. The immunology of hepatocellular carcinoma. Nat Immunol 2018; 19 (03) 222-232
- 4 Cho HJ, Cheong JY. Role of immune cells in patients with hepatitis B virus-related hepatocellular carcinoma. Int J Mol Sci 2021; 22 (15) 8011
- 5 Yu LX, Ling Y, Wang HY. Role of nonresolving inflammation in hepatocellular carcinoma development and progression. NPJ Precis Oncol 2018; 2 (01) 6
- 6 Hatziapostolou M, Polytarchou C, Aggelidou E. et al. An HNF4α-miRNA inflammatory feedback circuit regulates hepatocellular oncogenesis. Cell 2011; 147 (06) 1233-1247
- 7 Mínguez B, Hoshida Y, Villanueva A. et al. Gene-expression signature of vascular invasion in hepatocellular carcinoma. J Hepatol 2011; 55 (06) 1325-1331
- 8 Villanueva A, Newell P, Chiang DY, Friedman SL, Llovet JM. Genomics and signaling pathways in hepatocellular carcinoma. Semin Liver Dis 2007; 27 (01) 55-76
- 9 Poon D, Anderson BO, Chen LT. et al; Asian Oncology Summit. Management of hepatocellular carcinoma in Asia: consensus statement from the Asian Oncology Summit 2009. Lancet Oncol 2009; 10 (11) 1111-1118
- 10 Papatheodoridis GV, Lampertico P, Manolakopoulos S, Lok A. Incidence of hepatocellular carcinoma in chronic hepatitis B patients receiving nucleos(t)ide therapy: a systematic review. J Hepatol 2010; 53 (02) 348-356
- 11 Aravalli RN, Steer CJ, Cressman ENK. Molecular mechanisms of hepatocellular carcinoma. Hepatology 2008; 48 (06) 2047-2063
- 12 Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010; 140 (06) 883-899
- 13 Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011; 331 (6024) 1565-1570
- 14 Aravalli RN. Role of innate immunity in the development of hepatocellular carcinoma. World J Gastroenterol 2013; 19 (43) 7500-7514
- 15 Sachdeva M, Chawla YK, Arora SK. Immunology of hepatocellular carcinoma. World J Hepatol 2015; 7 (17) 2080-2090
- 16 Xue D, Zheng Y, Wen J. et al. Role of chemokines in hepatocellular carcinoma (Review). Oncol Rep 2021; 45 (03) 809-823
- 17 Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol 2017; 17 (09) 559-572
- 18 Kakinuma T, Hwang ST. Chemokines, chemokine receptors, and cancer metastasis. J Leukoc Biol 2006; 79 (04) 639-651
- 19 Makarova-Rusher OV, Medina-Echeverz J, Duffy AG, Greten TF. The yin and yang of evasion and immune activation in HCC. J Hepatol 2015; 62 (06) 1420-1429
- 20 Fu Y, Liu S, Zeng S, Shen H. From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma. J Exp Clin Cancer Res 2019; 38 (01) 396
- 21 Karin M. Nuclear factor-kappaB in cancer development and progression. Nature 2006; 441 (7092) 431-436
- 22 Cabillic F, Corlu A. Regulation of transdifferentiation and retrodifferentiation by inflammatory cytokines in hepatocellular carcinoma. Gastroenterology 2016; 151 (04) 607-615
- 23 Frau M, Biasi F, Feo F, Pascale RM. Prognostic markers and putative therapeutic targets for hepatocellular carcinoma. Mol Aspects Med 2010; 31 (02) 179-193
- 24 Silva MF, Sherman M. Criteria for liver transplantation for HCC: what should the limits be?. J Hepatol 2011; 55 (05) 1137-1147
- 25 Li J, Lau G, Chen L. et al. Interleukin 23 promotes hepatocellular carcinoma metastasis via NF-kappa B induced matrix metalloproteinase 9 expression. PLoS One 2012; 7 (09) e46264
- 26 Li XP, Yang XY, Biskup E. et al. Co-expression of CXCL8 and HIF-1α is associated with metastasis and poor prognosis in hepatocellular carcinoma. Oncotarget 2015; 6 (26) 22880-22889
- 27 Xue TC, Chen RX, Ren ZG, Zou JH, Tang ZY, Ye SL. Transmembrane receptor CXCR7 increases the risk of extrahepatic metastasis of relatively well-differentiated hepatocellular carcinoma through upregulation of osteopontin. Oncol Rep 2013; 30 (01) 105-110
- 28 Li B, Su H, Cao J, Zhang L. CXCL13 rather than IL-31 is a potential indicator in patients with hepatocellular carcinoma. Cytokine 2017; 89: 91-97
- 29 Lan X, Xiao F, Ding Q. et al. The effect of CXCL9 on the invasion ability of hepatocellular carcinoma through up-regulation of PREX2. J Mol Histol 2014; 45 (06) 689-696
- 30 Yang P, Li QJ, Feng Y. et al. TGF-β-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell 2012; 22 (03) 291-303
- 31 Li Y, Wang C, Yin X, Jiang L, Li X, Yang J. Profile and clinical significance of interferon gamma-inducible protein-10 (IP-10) and its receptor in patients with hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149 (16) 14879-14888
- 32 Kang Y, Su G, Sun J, Zhang Y. Activation of the TLR4/MyD88 signaling pathway contributes to the development of human hepatocellular carcinoma via upregulation of IL-23 and IL-17A. Oncol Lett 2018; 15 (06) 9647-9654
- 33 Gao S, Chen T, Li L. et al. Hypoxia-inducible ubiquitin specific peptidase 13 contributes to tumor growth and metastasis via enhancing the Toll-like receptor 4/myeloid differentiation primary response gene 88/nuclear factor-κB pathway in hepatocellular carcinoma. Front Cell Dev Biol 2020; 8: 587389
- 34 Zhang Y, Song T, Meng L, Wu X, Ba Y, Li Q. Melanoma antigen-1 mRNA combined with α-fetoprotein mRNA levels in peripheral blood of patients with hepatocellular carcinoma: a predictor of postoperative recurrence or metastasis?. ANZ J Surg 2009; 79 (1-2): 62-69
- 35 Targe M, Yasam VR, Nagarkar R. Hepatocellular carcinoma with uncommon sites of metastasis: a rare case report. Egypt J Radiol Nucl Med 2021; 52 (01) 228
- 36 Kummar S, Shafi NQ. Metastatic hepatocellular carcinoma. Clin Oncol (R Coll Radiol) 2003; 15 (05) 288-294
- 37 Matsui M, Machida S, Itani-Yohda T, Akatsuka T. Downregulation of the proteasome subunits, transporter, and antigen presentation in hepatocellular carcinoma, and their restoration by interferon-γ. J Gastroenterol Hepatol 2002; 17 (08) 897-907
- 38 Chan IH, Jain R, Tessmer MS. et al. Interleukin-23 is sufficient to induce rapid de novo gut tumorigenesis, independent of carcinogens, through activation of innate lymphoid cells. Mucosal Immunol 2014; 7 (04) 842-856
- 39 Kirchberger S, Royston DJ, Boulard O. et al. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J Exp Med 2013; 210 (05) 917-931
- 40 Liu Y, Song Y, Lin D. et al. NCR- group 3 innate lymphoid cells orchestrate IL-23/IL-17 axis to promote hepatocellular carcinoma development. EBioMedicine 2019; 41: 333-344
- 41 Warner K, Ghaedi M, Chung DC, Jacquelot N, Ohashi PS. Innate lymphoid cells in early tumor development. Front Immunol 2022; 13: 948358
- 42 Wu T, Yang W, Sun A, Wei Z, Lin Q. The role of CXC chemokines in cancer progression. Cancers (Basel) 2022; 15 (01) 167
- 43 Zajkowska M, Mroczko B. Chemokines in primary liver cancer. Int J Mol Sci 2022; 23 (16) 8846
- 44 Yin Z, Huang J, Ma T. et al. Macrophages activating chemokine (C-X-C motif) ligand 8/miR-17 cluster modulate hepatocellular carcinoma cell growth and metastasis. Am J Transl Res 2017; 9 (05) 2403-2411
- 45 Yu X, Chen Y, Cui L. et al. CXCL8, CXCL9, CXCL10, and CXCL11 as biomarkers of liver injury caused by chronic hepatitis B. Front Microbiol 2022; 13: 1052917
- 46 Kazanietz MG, Durando M, Cooke M. CXCL13 and its receptor CXCR5 in cancer: inflammation, immune response, and beyond. Front Endocrinol (Lausanne) 2019; 10: 471
- 47 Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm 2022; 3 (02) e147
- 48 Zhou C, Gao Y, Ding P, Wu T, Ji G. The role of CXCL family members in different diseases. Cell Death Discov 2023; 9 (01) 212
- 49 Zheng K, Li HY, Su XL. et al. Chemokine receptor CXCR7 regulates the invasion, angiogenesis and tumor growth of human hepatocellular carcinoma cells. J Exp Clin Cancer Res 2010; 29 (01) 31
- 50 Zhu F, Li X, Chen S, Zeng Q, Zhao Y, Luo F. Tumor-associated macrophage or chemokine ligand CCL17 positively regulates the tumorigenesis of hepatocellular carcinoma. Med Oncol 2016; 33 (02) 17
- 51 Li X, Yang G, Zhang W. et al. USP13: multiple functions and target inhibition. Front Cell Dev Biol 2022; 10: 875124
- 52 Huang J, Gu ZL, Chen W, Xu YY, Chen M. Knockdown of ubiquitin-specific peptidase 13 inhibits cell growth of hepatocellular carcinoma by reducing c-Myc expression. Kaohsiung J Med Sci 2020; 36 (08) 615-621
- 53 Li R, Gong J, Xiao C. et al. A comprehensive analysis of the MAGE family as prognostic and diagnostic markers for hepatocellular carcinoma. Genomics 2020; 112 (06) 5101-5114
- 54 Liu C, Xiao GQ, Yan LN. et al. Value of α-fetoprotein in association with clinicopathological features of hepatocellular carcinoma. World J Gastroenterol 2013; 19 (11) 1811-1819
- 55 Yoo DJ, Kim KM, Jin YJ. et al. Clinical outcome of 251 patients with extrahepatic metastasis at initial diagnosis of hepatocellular carcinoma: does transarterial chemoembolization improve survival in these patients?. J Gastroenterol Hepatol 2011; 26 (01) 145-154
- 56 Jun L, Zhenlin Y, Renyan G. et al. Independent factors and predictive score for extrahepatic metastasis of hepatocellular carcinoma following curative hepatectomy. Oncologist 2012; 17 (07) 963-969
- 57 Dang Y, Chen J, Feng W, Qiao C. et al. Interleukin 1β-mediated HOXC10 overexpression promotes hepatocellular carcinoma metastasis by upregulating PDPK1 and VASP. Theranostics 2020; 10 (08) 3833











