Abstract
Hepatocellular carcinoma (HCC) progresses through multiple stages of hepatocarcinogenesis, with each stage characterized by specific changes in vascular supply, drainage, and microvascular structure. These vascular changes significantly influence the imaging findings of HCC, enabling non-invasive diagnosis. Vascular changes in HCC are closely related to aggressive histological characteristics and treatment responses. Venous drainage from the tumor toward the portal vein in the surrounding liver facilitates vascular invasion, and the unique microvascular pattern of vessels that encapsulate the tumor cluster (known as a VETC pattern) promotes vascular invasion and metastasis. Systemic treatments for HCC, which are increasingly being used, primarily target angiogenesis and immune checkpoint pathways, which are closely intertwined. By understanding the complex relationship between histopathological vascular changes in hepatocarcinogenesis and their implications for imaging findings, radiologists can enhance the accuracy of imaging diagnosis and improve the prediction of prognosis and treatment response. This, in turn, will ultimately lead to better patient care.
Keywords: Hepatocellular carcinoma, Hepatocarcinogenesis, Angiogenesis, Vascular invasion, Systemic therapy
INTRODUCTION
Liver cancer is the sixth most prevalent cancer worldwide and the third most common cause of cancer-related deaths in 2020. The incidence of and the number of deaths from liver cancer are anticipated to increase over the next 20 years [1,2]. Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and accounts for approximately 80% of all primary liver cancers [3].
Most contemporary guidelines recommend diagnosing HCC in high-risk patients using radiological hallmarks. These hallmarks are characterized by arterial phase hyperenhancement and washout and are based on histopathological changes in the vascular supply that occur during hepatocarcinogenesis [4,5,6]. During hepatocarcinogenesis, changes in vascular drainage contribute to microscopic and macroscopic vascular invasion, which is a significant determinant of treatment decisions and a well-established prognostic indicator [7]. Moreover, HCC treatment depends significantly on its vascular characteristics. Utilizing the dependence of HCC on the hepatic artery for blood supply, transarterial therapies such as transarterial chemoembolization and transarterial radioembolization have been employed [8,9,10]. In addition, HCC often exhibits high expression of angiogenic factors, such as vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang2), leading to the frequent use of therapies targeting angiogenesis in systemic treatment [7,11]. Therefore, understanding the stepwise vascular changes that occur during the progression of hepatocarcinogenesis is crucial for diagnosing HCC, predicting prognosis, and determining treatment strategies [12].
In this review article, we explored the histopathological vascular evolution associated with hepatocarcinogenesis and its implications for imaging-based diagnosis, treatment, and prognosis of HCC.
Vascular Changes Associated With HCC Progression
HCC progresses in multiple steps, each characterized by distinctive histological and molecular changes. Histologically, progression follows a sequence from 1) regenerative nodules in a cirrhotic liver, 2) precancerous lesions, such as dysplastic nodules (DNs) or dysplastic foci, 3) early HCC, and 4) progressed HCC. It is important to note that progressive HCC is not a homogeneous group of tumors; instead, it exhibits various histological and molecular characteristics. In this review, we categorized progressed HCC into two stages based on tumor differentiation: moderately differentiated HCC (MD-HCC) and poorly differentiated HCC (PD-HCC). The vascular changes that occur at each stage of HCC are summarized in Figure 1.
Fig. 1. Summary of vascular changes and imaging implications in each stage of hepatocarcinogenesis. LC = liver cirrhosis, DN = dysplastic nodule, HCC = hepatocellular carcinoma, MD = moderately differentiated, PD = poorly differentiated, PV = portal vein, VETC = vessels that encapsulate tumor cluster.
Vascular System of Liver
The liver is nourished by a dual blood supply: the portal venous system, which delivers oxygen and nutrients, and the hepatic arteries. The portal vein supplies venous blood to the liver from the gastrointestinal tract and the spleen [13]. Meanwhile, the hepatic artery, portal vein, and bile duct run parallel to one another and are collectively referred to as the portal triad. Microscopically, the hepatic lobule is a basic structural unit of the liver. The hepatic vein lies at the center of the hepatic lobule and is surrounded by multiple portal triads that form a polygonal shape (Supplementary Fig. 1). Sinusoids, the liver microvasculature, connect the portal triad to the hepatic vein. Sinusoids are lined by liver sinusoidal endothelial cells, which are characterized by the absence of a basement membrane and the presence of fenestrations that facilitate the exchange of macromolecules between hepatocytes and blood [14].
Early Hepatocarcinogenesis Up to the Early HCC Stage
In early HCC, tumor cells acquire invasiveness and penetrate and destroy internal structures within the lesion, including hepatic veins, portal tracts, and fibrous septa [15]. This invasive growth of tumor cells is called stromal invasion and serves as histopathological evidence that early HCC is a malignant tumor [16]. Notably, as the intralesional hepatic vein lacks the surrounding fibrosis present around the portal triad, it is more vulnerable to destruction by stromal invasion. Consequently, as the number of intralesional hepatic veins within a lesion decreases, venous drainage from the lesion is redirected toward either the hepatic sinusoid or portal vein [17].
The progression of stromal invasion leads to the gradual destruction of the portal triad following the hepatic vein. Destruction of the portal triad causes a decrease in vascular supply, leading to intralesional hypoxia [18]. Hypoxia stimulates angiogenic signaling, including VEGF, which results in the growth of new arterial branches in HCC [19,20]. Unlike the original portal tracts, these newly developed arteries do not accompany the portal vein or bile duct and are thus referred to as unpaired arteries. The development of unpaired arteries begins in the early HCC stage and continues until MD-HCC. Therefore, it is common for the vascularity of a lesion to be lower than or similar to that of the surrounding non-tumor liver tissue during the early HCC stage [21]. Fatty changes occurring alongside a decrease in HCC vascularity are observed in approximately 40% of early HCC cases, particularly in lesions measuring 1 and 1.5 cm. At this stage, fatty changes often diffused throughout the lesion [18]. Although the lack of blood supply and increased cell density may cause intralesional hypoxia and fatty change, the molecular mechanism between the intralesional hypoxia and diffuse fatty changes remains elusive.
Sinusoidal capillarization refers to the acquisition of a capillary-like phenotype by liver sinusoidal endothelial cells, which includes the loss of fenestrae, accumulation of basement membranes, and alterations in cell-cell and cell-matrix interactions [22]. With sinusoidal capillarization, the expression of adhesion markers such as CD31 and CD34 increases, and these can be used to assess the distribution or density of capillarized sinusoids using immunohistochemistry [23]. Sinusoidal capillarization begins as a phenomenon of fibrogenesis and sinusoidal remodeling during liver cirrhosis, accelerates at the high-grade DN stage, and progresses rapidly to the early HCC and MD-HCC stages [20,24]. As sinusoidal capillarization is a gradual process, the degree of capillarization is not used to distinguish between early HCC and high-grade DNs [20,22,25].
Collectively, during the early HCC stage, tumor invasiveness triggers vascular changes. This stage is characterized by the destruction of preexisting vascular structures, accompanied by incomplete neoangiogenesis and sinusoidal capillarization. Owing to incomplete neoangiogenesis, arterial hyperenhancement is not commonly observed on CT or MRI in early HCC (Fig. 2). Additionally, only a small proportion (0%–31%) of early HCC exhibits arterial hyperenhancement [21,26,27]. Because early HCC does not show arterial hyperenhancement, it is challenging to diagnose using the diagnostic hallmarks of HCC [27]. Changes in vascular supply and drainage during this period are better visualized with CT during hepatic arteriography (CTHA) and CT during arterial portography (CTAP). In CTHA and CTAP, contrast media are injected into the hepatic artery and superior mesenteric artery through a catheter, respectively, allowing separate assessments of arterial and portal blood supplies [28]. Using these methods, it can be observed that both arterial and portal blood supplies are reduced in the early HCC stage [17].
Fig. 2. A 53-year-old male with alcoholic hepatitis and early HCC. A-F: Gadoxetic acid-enhanced MRI revealed an approximately 1.5-cm HCC in the right posterior liver (arrows). The lesion exhibits isointensity on the pre-contrast T1-weighted image (A), faint and partial hyperenhancement on the arterial phase (B), absence of washout on the portal phase (C), hypointensity on the hepatobiliary phase (D), mild hyperintensity on the diffusion-weighted image (b = 800) (E), and isointensity on the T2-weighted image (F). G: The lesion was identified as early HCC and displayed a vaguely nodular appearance in the gross specimen (arrows). H: A small number of unpaired arteries (arrowheads) were observed within the lesion (α-smooth muscle actin staining, ×100). I: Compared to the surrounding non-tumor liver (left side), partial sinusoidal capillarization occurred in the early HCC stage (right side) (CD34 staining, ×100). HCC = hepatocellular carcinoma.
Ancillary features such as high signal intensity on diffusion-weighted or T2-weighted images and hypointensity in the hepatobiliary phase can assist in differentiating early HCC from DNs [26,27]. A signal drop in the T1-opposed phase is also observed in 26%–53% of early HCC cases due to fatty changes [21,26]. However, fatty changes in HCC are not only limited to the early HCC stage but are also commonly observed in progressive HCC accompanied by fatty liver disease and metabolic syndrome [29].
Progressed HCC: MD-HCC
At this stage, the vascular changes that begin during early hepatocarcinogenesis are completed. The portal triad nearly disappears and, to compensate, the number of unpaired arteries significantly increases [20,22]. As a result, the HCC becomes almost entirely dependent on unpaired arteries for its vascular supply, resulting in hyperenhancement during the arterial phase compared to the non-tumor liver, which mainly relies on the portal vein for vascular supply [30,31]. Notably, the degree of arterial enhancement is significantly correlated with the density of unpaired arteries (Fig. 3) [32].
Fig. 3. A 61-year-old male with C-viral hepatitis and moderately differentiated HCC. A-C: CT revealed an approximately 2-cm HCC in the dome of the right liver. The lesion demonstrates isoattenuation on the pre-contrast image (A), hyperenhancement in the arterial phase (B), and an enhancing capsule and equivocal washout in the portal phase (C). D: The lesion was identified as moderately differentiated-HCC and exhibited high density of unpaired arteries (arrowheads) within the lesion (α-smooth muscle actin staining, ×200). E: Compared to the surrounding non-tumor liver (left side), completely capillarized capillary pattern microvessels were observed in the HCC (right side) (CD34 staining, ×100). HCC = hepatocellular carcinoma.
During the portal phase, the contrast media from the portal blood flow enhances the non-tumor liver, while HCC is not well enhanced because the contrast media that enters during the arterial phase exits, and there is limited incoming portal blood flow. This results in the relative hypoenhancement of HCC, known as washout. Other factors contributing to washout include high tumor cellularity with a small extracellular volume, expanded extracellular space of the fibrotic liver surrounding the tumor, and intrinsic hypoattenuation or hypointensity [33]. Many HCCs also exhibit a washout appearance in the delayed phase. However, the mechanism of washout observed in the delayed phase is not fully understood, although recent studies have indicated that variations in the extracellular volume percentage are significant [34].
Sinusoidal capillarization is observed in the predominant areas of most HCCs at this MD-HCC stage [20]. Notably, the density of capillarized sinusoids, often referred to as microvascular density (MVD), is associated with arterial phase hyperenhancement [35,36]. Additionally, as sinusoidal capillarization occurs in parallel with neoangiogenesis, it is uncertain whether their relationship is causal or merely correlative [20].
The hepatic vein nearly disappears in MD-HCC, and venous drainage occurs from the intratumoral sinusoids through the capsule to the peritumoral portal vein [17]. As a result, the area drained from the HCC is typically clearly visible on CTHA. As intrahepatic metastasis occurs primarily in this area, it can be utilized in planning surgical resections [37]. On dynamic CT and MRI, this phenomenon appears as a corona enhancement. However, unlike when employing CTHA, the observation of corona enhancement is limited due to the enhancement of the non-tumor liver in the portal phase.
Progressed HCC: PD-HCC Stage
The density of unpaired arteries in HCC, which is responsible for the intratumoral blood supply, is lower in PD-HCC than in MD-HCC [12,20,38]. Since unpaired artery density is closely associated with arterial hyperenhancement in HCC, a decrease in unpaired artery density leads to decreased arterial hyperenhancement [12,38]. However, the mechanism underlying the decrease in the density of unpaired arteries in PD-HCC is not well understood. A previous study suggested that the acquisition of hypervascularity at the MD-HCC stage led to a reduction in VEGF expression, resulting in decreased vascularity at the subsequent PD-HCC stage [38]. Notably, in the PD-HCC stage, the majority of cases display a heterogeneous enhancement pattern with a mix of hypervascular and hypovascular portions [38,39]. Furthermore, some tumors exhibit central hypovascularity with hyperenhancement in the outer rim, often referred to as rim-like enhancement (Fig. 4). In these cases, arterial phase rim enhancement is associated with various histological characteristics, including the macrotrabecular-massive (MTM) subtypes, fibrosis, and necrosis [40]. These features are believed to be additional factors contributing to the hypovascularity of PD-HCC.
Fig. 4. A 57-year-old male with B-viral hepatitis and poorly differentiated HCC. A-F: Gadoxetic acid–enhanced MRI revealed an approximately 4-cm HCC in liver segment 5. The lesion exhibits hypointensity in the pre-contrast T1-weighted image (A), rim enhancement in the arterial phase (B), hypointensity in the portal phase (C) and hepatobiliary phase (D), hyperintensity in the diffusion-weighted image (b = 800) (E), and hyperintensity on the T2-weighted image (F). G: The lesion was identified as poorly differentiated-HCC and displayed vessels that encapsulate the tumor cluster pattern and macrotrabecular patterns (CD34 staining, ±100). HCC = hepatocellular carcinoma.
As HCC progresses, the pattern and density of intratumoral sinusoidal vessels also change. Sinusoidal vessels can be classified into two patterns: a capillary pattern characterized by small, scattered microvessels with no or narrow lumens and a sinusoidal pattern featuring continuous branching and an obvious lumen structure [41]. The capillary pattern is more common in well-differentiated HCC (WD-HCC) and MD-HCC than in PD-HCC, whereas the incidence of a sinusoidal pattern progressively increases with HCC progression. Microvessels in the sinusoidal pattern are also associated with a lower MVD, and PD-HCC has been reported to have a lower MVD than MD-HCC [42,43]. Collectively, both unpaired arterial density and MVD decrease in PD-HCC, leading to reduced arterial hyperenhancement and more frequent tumor necrosis [12,43].
VETC Pattern: A Peculiar Microvascular Pattern Common in the Late Stage of Hepatocarcinogenesis
Recently, Fang et al. [44] described microvessels in a sinusoidal pattern that encapsulated and separated tumor clusters as “vessels that encapsulate the tumor cluster (VETC)” pattern. This is a specific type of sinusoidal pattern, and it is important to note that other sinusoidal patterns may not qualify as VETC patterns [45]. HCC with a VETC pattern (VETC-HCC) is commonly defined as HCC displaying any (≥1%–5%) VETC pattern within the tumor [44,46,47]. However, some studies recommend using a cutoff of 50%–55% of the tumor area for VETC, which is considered optimal for predicting prognosis [48,49]. The reported incidence of VETC-HCC varies widely, ranging from 22.0%–76.5% of HCC cases with a 1%–5% VETC area cutoff and 2.9%–36.5% with a 50%–55% cutoff [50].
Traditionally, the epithelial-mesenchymal transition (EMT) has been considered a crucial step in metastasis and is characterized by decreased cell-to-cell adhesion and increased migration and invasion, allowing cancer cells to enter the bloodstream [51]. However, the endothelium-encapsulated tumor clusters of VETC-HCC can directly enter the bloodstream without undergoing the complex invasion process associated with EMT (Fig. 5) [44,52]. Therefore, VETC-HCC can form tumor microemboli at intrahepatic and extrahepatic sites in an EMT-independent manner, frequently resulting in hematogenous spread, particularly to the lungs [48,53]. Additionally, VETC-HCC is associated with larger tumor sizes (>5 cm), more advanced stages, higher levels of serum alpha-fetoprotein and protein induced by vitamin K antagonist-II, and poorer differentiation [44,46,47,48].
Fig. 5. Schematic representation of two types of metastases of HCC. HCC = hepatocellular carcinoma, VETC = vessels that encapsulate tumor clusters, EMT = epithelial-mesenchymal transition.

VETC-HCC is frequently associated with MTM-HCC [48,54,55]. MTM-HCC is defined as HCC with a macrotrabecular pattern characterized by trabeculae thicker than six cells, predominating more than 50% of the tumor area [56]. MTM-HCC often features microvessels with a sinusoidal pattern, including VETC, and commonly shows the expression of hypoxia markers and necrosis [40,57]. Intralesional hypoxia and necrosis are believed to occur in central area of thick trabeculae that are located far from the vascular spaces, leading to insufficient oxygen supply. Due to its connection with several aggressive clinical and histological characteristics, VETC-HCC is linked to a poor prognosis following resection or transplantation [48,54].
The dynamic CT and MRI features of VETC-HCC are characterized by large tumor size (>5 cm), irregular rim-like enhancement in the arterial phase, and intratumoral necrosis (Fig. 4) [40,58,59]. Hypovascularity in VETC-HCC is speculated to arise from low MVD in HCCs exhibiting a sinusoidal pattern [40]. The pathological, clinical, and imaging characteristics of VETC-HCC are summarized in Table 1.
Table 1. Pathological, clinical, and imaging characteristics of VETC-HCC.
| Pathological characteristics | |
| Micro and/or microvascular invasion | |
| Poor differentiation | |
| Macrotrabecular-massive subtype | |
| Clinical characteristics | |
| Large tumor size | |
| Advanced stage | |
| High level of serum AFP and PIVKA-II | |
| Imaging characteristics | |
| Large tumor size | |
| Irregular rim-like enhancement in the arterial phase | |
| Intratumoral necrosis | |
VETC-HCC = hepatocellular carcinoma with vessels that encapsulate tumor cluster pattern, AFP = alpha-fetoprotein, PIVKA-II = protein induced by vitamin K antagonist II
A previous report indicates that Ang2 is crucial for the formation of the VETC pattern. In a xenograft model of VETC-HCC, silencing Ang2 expression suppressed the VETC pattern and reduced metastasis [44]. Furthermore, HCC tissues with VETC patterns tend to exhibit high Ang2 expression [47,55]. Ang2, a proangiogenic factor in the angiopoietin/TIE2 pathway, plays a vital role in HCC angiogenesis and functions synergistically with the VEGF pathway [60]. Similar to the prevalence of HCCs with a VETC pattern in the advanced stages, the expression of Ang2 in HCC tissues typically occurs later than VEGF expression [61,62]. Notably, high Ang2 mRNA or protein expression in HCC tissues and high serum Ang2 levels are associated with poor prognoses [62,63,64].
Micro- and Macrovascular Invasion: HCC Dissemination via Altered Vascular Drainage
HCC commonly exhibits microvascular invasion (MiVI) and/or macrovascular invasion (MaVI), which indicate aggressive behavior, metastatic potential, and poor prognosis [7]. MiVI is defined as the presence of tumor emboli in the microscopic vessels of the tumor capsule or adjacent non-tumor liver [65]. However, the specific definition of MiVI differs significantly among studies, particularly in terms of the types of microvessels and their distance from the HCC where MiVI is observed [65].
MiVI can be observed in small, thin-walled vessels in the adjacent liver tissue or thicker-walled vessels such as the portal vein or hepatic vein. Some studies have also included the presence of tumor cells in the hepatic artery or lymphatic vessels as part of MiVI [65,66]. The reported prevalence of MiVI ranges widely from 15%–57% in surgically treated HCC, which is thought to reflect the variation in the stages of HCC included in the studies and the diversity in the definitions used [65]. Moreover, there have been attempts to further categorize MiVI pathologically, with reports indicating that the prognosis tends to be worse when MiVI is observed in multiple locations, involves a large number of invaded tumor cells, or includes invasion of microscopic portal veins [66,67].
MiVI is believed to result from tumor emboli originating from HCC that disseminate along the paths of blood drainage [68,69]. In progressed HCC, the drainage routes are primarily through the capsular microvessels and portal vein of the adjacent non-tumor liver, where MiVI commonly occurs [68]. Consequently, MiVI is believed to develop into a gross vascular invasion or intrahepatic metastasis. The fact that progressed HCC rarely drains directly into the hepatic vein might explain why HCC recurrence is much more common at intrahepatic sites than at extrahepatic sites.
MiVI is a widely recognized prognostic factor for HCC and is associated with poor disease-free and overall survival after hepatic resection and liver transplantation [65]. It is included as a poor prognostic factor in the 5th World Health Organization classification and 8th edition of the American Joint Committee on Cancer (AJCC) tumor-node-metastasis (TNM) staging system [70,71]. One drawback of using MiVI as a prognostic factor is that it requires microscopic evaluation of the peritumoral area, meaning that it can only be identified after surgical treatment has been completed and cannot be assessed through biopsy before treatment. To overcome this limitation, numerous studies have aimed to predict MiVI non-invasively through imaging [72]. Imaging findings that indicate aggressive histopathological characteristics of HCC, such as large tumor size (>5 cm), multifocality, arterial phase rim enhancement, and non-smooth tumor margins, have been reported to be associated with MiVI (Fig. 6) [39,72]. Imaging features such as arterial phase peritumoral enhancement and peritumoral hypointensity in the hepatobiliary phase are also linked to MiVI and are thought to reflect peritumoral perfusion abnormalities caused by MiVI [39]. These imaging findings can be evaluated before treatment, potentially aiding in deciding between surgical resection and ablation or in assessing candidates for liver transplantation in the management of HCC [73,74,75].
Fig. 6. A 54-year-old man with moderately differentiated HCC with microvascular invasion and a satellite nodule. A-F: Gadoxetic acid-enhanced MRI revealed an approximately 3-cm HCC in liver segment 5/6. The lesion exhibits hypointensity on the pre-contrast T1-weighted image (A), hyperenhancement in the arterial phase (B), hypointensity in the portal phase (C) and hepatobiliary phase (D), hyperintensity in the diffusion-weighted image (b = 800) (E), and hyperintensity on the T2-weighted image (F). The lesion shows arterial phase peritumoral enhancement and peritumoral hypointensity in the hepatobiliary phase (arrows) and a non-smooth tumor margin in the portal and hepatobiliary phases, suggesting a high risk of microvascular invasion. A satellite nodule is observed in the area where the peritumoral change occurred (arrowheads). HCC = hepatocellular carcinoma.
MaVI is defined as the presence of a tumor in a major vessel that can be identified during macroscopic examination or imaging. MaVI is also a well-known poor prognostic factor and is part of established staging systems such as the Barcelona Clinic Liver Cancer (BCLC) classification, AJCC TNM stage, and selection criteria for liver transplantation [7,71,76]. The presence of MaVI not only affects tumor staging but is also crucial in making treatment decisions [77]. In HCC, MaVI predominantly presents as tumor thrombosis within the portal or hepatic veins. Although a hepatic mass accompanied by tumor in vein (TIV) often suggests HCC, a similar appearance can occur in intrahepatic cholangiocarcinoma (CCA) or combined HCC-CCA [78]. Among HCC patients, TIV is observed in 7%–60% of cases and is especially common in HCC displaying an infiltrative appearance, where it is seen in 68%–100% of such cases [77,79,80]. In instances where infiltrative HCC, it can mimic cirrhotic nodules, displaying a “cirrhotomimetic” appearance. Therefore, the presence of TIV can serve as a useful diagnostic clue for infiltrative HCC [79].
In patients with HCC, bland thrombosis may arise from portal hypertension or venous obstruction caused by the HCC [77]. Therefore, distinguishing between branch thrombosis and TIV is clinically important. Key CT and MRI features for differentiating TIV from bland thrombosis include unequivocal enhancement, an ill-defined venous wall, contiguity with the hepatic mass, restricted diffusion, shunting to the intravenous space, and signal intensity similar to that of the hepatic mass [77,81]. Notably, CT and MRI display a sensitivity of 62%–93% and specificity of 87%–100% for TIV. Overall, although their diagnostic accuracies are similar, MRI is known to have a marginally better sensitivity [82,83,84,85].
Systemic Treatment: Implication of Angiogenic Factors and Immunovascular Phenotype
Systemic therapy is recommended for advanced-stage (BCLC stage C) HCC. Sorafenib was the first systemic agent approved for HCC, and lenvatinib was later approved as a first-line drug after demonstrating non-inferiority to sorafenib [86,87]. Regorafenib serves as a second-line treatment, offering survival benefits to patients who show disease progression after sorafenib therapy [88]. HCC, characterized by its hypervascular nature, exhibits a high expression of angiogenic factors. The approved therapeutic agents target a variety of angiogenic factors: sorafenib targets VEGFR1-3 and PDGFR-β; lenvatinib targets VEGFR1-3, FGFR1-4, and PDGFR-α; regorafenib targets VEGFR1-3, PDGFR-β, FGFR1, and TIE2 [89]. Other second-line agents such as ramucirumab and cabozantinib also inhibit angiogenic factors such as VEGFR2 or TIE2 [89,90].
The scheme of systemic therapy for HCC has recently shifted toward immunotherapy, with atezolizumab-bevacizumab and durvalumab-tremelimumab combination therapies recommended as first-line drugs and sorafenib, lenvatinib, or durvalumab monotherapies suggested only when the former are not feasible [7]. Atezolizumab is an anti-PD-L1 antibody and immune checkpoint inhibitor, whereas bevacizumab is an anti-VEGFA antibody that targets the angiogenic factor VEGFA [90]. The combination of atezolizumab-bevacizumab not only provides anti-angiogenic effects but also enhances anticancer immunity in the immunosuppressed tumor microenvironment of HCC, creating synergy with atezolizumab [91]. In contrast, durvalumab and tremelimumab, which target anti-PD-L1 and anti-CTLA4 respectively, are immune checkpoint inhibitors [90,92]. Collectively, antiangiogenic therapy and immunotherapy are the two main types of systemic therapies for HCC (Fig. 7).
Fig. 7. Inhibition of angiogenic and immune checkpoint pathways by systemic therapeutic agents for HCC. HCC = hepatocellular carcinoma, CTLA4 = cytotoxic T-lymphocyte associated protein 4, PD1 = programmed cell death protein 1, PD-L1 = programmed cell death-ligand 1, VEGFA = vascular endothelial growth factor A, FGFR = fibroblast growth factor receptor, PDGFR = platelet-derived growth factor receptor, VEGFR = vascular endothelial growth factor receptor.
Interestingly, the vascular and immune phenotypes within the HCC microenvironment are closely intertwined, allowing classification into four groups: immune-high/angiostatic, immune-mid/angio-mid, immune-low/angiogenic, and immune-low/angio-low phenotypes [55]. The immune-high/angiostatic group is characterized by high lymphocyte infiltration and low expression of angiogenic factors, whereas the immune-low/angiogenic group exhibits low lymphocyte infiltration, high expression of angiogenic factors, histologic patterns of VETC and MTM, and frequent mutations in CTNNB1. Understanding the pathological and molecular diversity of HCC is crucial for determining the optimal sequence for systemic therapies. Indeed, VETC-HCC, recognized for its high expression of angiogenic factors, benefits from sorafenib treatment, whereas non-VETC-HCC does not [46]. Another report suggested that HCC with arterial phase rim enhancement, which is known to have aggressive clinicopathological characteristics, showed a more favorable response to atezolizumab-bevacizumab treatment [93]. However, research on therapeutic tailoring based on pathological, molecular, and imaging phenotypes of HCC is limited.
Summary and Future Perspectives
Vascular changes in HCC represent a complex, multistep process that occurs during hepatocarcinogenesis. The reliance of HCC on the hepatic artery for vascular supply is its primary cause of its diagnostic hallmark. In the early stages of HCC, typical imaging characteristics are often absent due to immature changes in the vascular supply. Hypovascularity and fatty acid changes are also observed more frequently. In the PD-HCC stage, dependence on the arterial vascular supply decreases while microvessels transform into a sinusoidal pattern. VETC-HCC, often emerging in the PD-HCC stage, displays atypical imaging features such as large tumor size, arterial phase rim enhancement, and necrosis. In HCC, along with changes in the vascular supply, there are also alterations in vascular drainage, with drainage transitioning from the hepatic vein to the portal vein of the surrounding non-tumor liver. Following these altered vascular drainage routes, tumor emboli frequently occur in the small vessels of the adjacent non-tumorous liver, a phenomenon known as MiVI. MiVI can progress to MaVI or intrahepatic metastasis.
The vascular changes that occur during multistep hepatocarcinogenesis lead to various imaging findings in HCC. The most widely used application of these imaging findings in clinical practice is the diagnosis of HCC [4,5,6]. Additionally, features such as fatty changes, commonly observed in early HCC, and corona enhancement, observed in progressed HCC, are also recommended as ancillary diagnostic features.
VETC-HCC and MiVI, which are commonly observed in the late stages of hepatocarcinogenesis, are well-known poor prognostic factors, and their associated imaging findings can be useful for predicting HCC prognosis [39,72]. Previous studies have suggested that these imaging findings are useful for the prognostic prediction of pathological findings [94]. However, most of these studies were retrospective, and multicenter prospective evidence is lacking. Another issue is that the evaluation of these imaging findings by radiologists is often subjective, leading to low inter-reader agreement [95].
Systemic therapy is increasingly used to treat HCC, with the most approved systemic agents targeting angiogenic or immune checkpoint pathways. Interestingly, the vascular phenotype and immune microenvironment of HCC are closely related, suggesting that imaging findings may predict the response to systemic therapy. Even when treated with atezolizumab-bevacizumab, the current first-line agent for HCC, 19%–37% of patients show no response and experience progressive disease after the first cycle [93,96,97]. As most systemic agents for HCC are costly, a reliable imaging biomarker for response prediction and optimization of the sequence of systemic agents is urgently needed for HCC treatment.
In conclusion, by understanding the intricate connections between histopathological vascular evolution and its implications for imaging findings in hepatocarcinogenesis, radiologists can enhance diagnostic accuracy and prognostic prediction, ultimately improving patient care.
Acknowledgments
Illustrations (Fig. 5, Fig. 7, Supplementary Fig. 1A) were created with BioRender.com.
Footnotes
Conflicts of Interest: The authors have no potential conflicts of interest to disclose.
- Funding acquisition: Hyungjin Rhee.
- Supervision: Jin-Young Choi.
- Writing—original draft: all authors.
- Writing—review & editing: all authors.
Funding Statement: This work was supported by the National Research Foundation of Korea (NRF) grant, funded by the Korean government (MSIT) (No. RS-2023-00208307).
Supplement
The Supplement is available with this article at https://doi.org/10.3348/kjr.2024.0307.
References
- 1.Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77:1598–1606. doi: 10.1016/j.jhep.2022.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kim DY. Changing etiology and epidemiology of hepatocellular carcinoma: Asia and worldwide. J Liver Cancer. 2024;24:62–70. doi: 10.17998/jlc.2024.03.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rumgay H, Ferlay J, de Martel C, Georges D, Ibrahim AS, Zheng R, et al. Global, regional and national burden of primary liver cancer by subtype. Eur J Cancer. 2022;161:108–118. doi: 10.1016/j.ejca.2021.11.023. [DOI] [PubMed] [Google Scholar]
- 4.Korean Liver Cancer Association (KLCA) and National Cancer Center (NCC) Korea. 2022 KLCA-NCC Korea practice guidelines for the management of hepatocellular carcinoma. Korean J Radiol. 2022;23:1126–1240. doi: 10.3348/kjr.2022.0822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Singal AG, Llovet JM, Yarchoan M, Mehta N, Heimbach JK, Dawson LA, et al. AASLD practice guidance on prevention, diagnosis, and treatment of hepatocellular carcinoma. Hepatology. 2023;78:1922–1965. doi: 10.1097/HEP.0000000000000466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.European Association for the Study of the Liver. EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2018;69:182–236. doi: 10.1016/j.jhep.2018.03.019. [DOI] [PubMed] [Google Scholar]
- 7.Reig M, Forner A, Rimola J, Ferrer-Fàbrega J, Burrel M, Garcia-Criado Á, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76:681–693. doi: 10.1016/j.jhep.2021.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sangro B, Salem R. Transarterial chemoembolization and radioembolization. Semin Liver Dis. 2014;34:435–443. doi: 10.1055/s-0034-1394142. [DOI] [PubMed] [Google Scholar]
- 9.Cho Y, Choi JW, Kwon H, Kim KY, Lee BC, Chu HH, et al. Transarterial chemoembolization for hepatocellular carcinoma: 2023 expert consensus-based practical recommendations of the Korean Liver Cancer Association. Korean J Radiol. 2023;24:606–625. doi: 10.3348/kjr.2023.0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Choi JW, Kim HC. Radioembolization for hepatocellular carcinoma: what clinicians need to know. J Liver Cancer. 2022;22:4–13. doi: 10.17998/jlc.2022.01.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bruix J, Chan SL, Galle PR, Rimassa L, Sangro B. Systemic treatment of hepatocellular carcinoma: an EASL position paper. J Hepatol. 2021;75:960–974. doi: 10.1016/j.jhep.2021.07.004. [DOI] [PubMed] [Google Scholar]
- 12.Asayama Y, Yoshimitsu K, Nishihara Y, Irie H, Aishima S, Taketomi A, et al. Arterial blood supply of hepatocellular carcinoma and histologic grading: radiologic-pathologic correlation. AJR Am J Roentgenol. 2008;190:W28–W34. doi: 10.2214/AJR.07.2117. [DOI] [PubMed] [Google Scholar]
- 13.Carneiro C, Brito J, Bilreiro C, Barros M, Bahia C, Santiago I, et al. All about portal vein: a pictorial display to anatomy, variants and physiopathology. Insights Imaging. 2019;10:38. doi: 10.1186/s13244-019-0716-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D, et al. Liver sinusoidal endothelial cells: physiology and role in liver diseases. J Hepatol. 2017;66:212–227. doi: 10.1016/j.jhep.2016.07.009. [DOI] [PubMed] [Google Scholar]
- 15.Kondo F. Assessment of stromal invasion for correct histological diagnosis of early hepatocellular carcinoma. Int J Hepatol. 2011;2011:241652. doi: 10.4061/2011/241652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.International Consensus Group for Hepatocellular Neoplasia. Pathologic diagnosis of early hepatocellular carcinoma: a report of the international consensus group for hepatocellular neoplasia. Hepatology. 2009;49:658–664. doi: 10.1002/hep.22709. [DOI] [PubMed] [Google Scholar]
- 17.Kitao A, Zen Y, Matsui O, Gabata T, Nakanuma Y. Hepatocarcinogenesis: multistep changes of drainage vessels at CT during arterial portography and hepatic arteriography--radiologic-pathologic correlation. Radiology. 2009;252:605–614. doi: 10.1148/radiol.2522081414. [DOI] [PubMed] [Google Scholar]
- 18.Kutami R, Nakashima Y, Nakashima O, Shiota K, Kojiro M. Pathomorphologic study on the mechanism of fatty change in small hepatocellular carcinoma of humans. J Hepatol. 2000;33:282–289. doi: 10.1016/s0168-8278(00)80369-4. [DOI] [PubMed] [Google Scholar]
- 19.Yang ZF, Poon RT. Vascular changes in hepatocellular carcinoma. Anat Rec (Hoboken) 2008;291:721–734. doi: 10.1002/ar.20668. [DOI] [PubMed] [Google Scholar]
- 20.Park YN, Kim YB, Yang KM, Park C. Increased expression of vascular endothelial growth factor and angiogenesis in the early stage of multistep hepatocarcinogenesis. Arch Pathol Lab Med. 2000;124:1061–1065. doi: 10.5858/2000-124-1061-IEOVEG. [DOI] [PubMed] [Google Scholar]
- 21.Rhee H, Kim MJ, Park YN, Choi JS, Kim KS. Gadoxetic acid-enhanced MRI findings of early hepatocellular carcinoma as defined by new histologic criteria. J Magn Reson Imaging. 2012;35:393–398. doi: 10.1002/jmri.22828. [DOI] [PubMed] [Google Scholar]
- 22.Park YN, Yang CP, Fernandez GJ, Cubukcu O, Thung SN, Theise ND. Neoangiogenesis and sinusoidal “capillarization” in dysplastic nodules of the liver. Am J Surg Pathol. 1998;22:656–662. doi: 10.1097/00000478-199806000-00002. [DOI] [PubMed] [Google Scholar]
- 23.Couvelard A, Scoazec JY, Feldmann G. Expression of cell-cell and cell-matrix adhesion proteins by sinusoidal endothelial cells in the normal and cirrhotic human liver. Am J Pathol. 1993;143:738–752. [PMC free article] [PubMed] [Google Scholar]
- 24.Thabut D, Shah V. Intrahepatic angiogenesis and sinusoidal remodeling in chronic liver disease: new targets for the treatment of portal hypertension? J Hepatol. 2010;53:976–980. doi: 10.1016/j.jhep.2010.07.004. [DOI] [PubMed] [Google Scholar]
- 25.Park YN. Update on precursor and early lesions of hepatocellular carcinomas. Arch Pathol Lab Med. 2011;135:704–715. doi: 10.5858/2010-0524-RA.1. [DOI] [PubMed] [Google Scholar]
- 26.Sano K, Ichikawa T, Motosugi U, Sou H, Muhi AM, Matsuda M, et al. Imaging study of early hepatocellular carcinoma: usefulness of gadoxetic acid-enhanced MR imaging. Radiology. 2011;261:834–844. doi: 10.1148/radiol.11101840. [DOI] [PubMed] [Google Scholar]
- 27.Rhee H, Kim MJ, Park MS, Kim KA. Differentiation of early hepatocellular carcinoma from benign hepatocellular nodules on gadoxetic acid-enhanced MRI. Br J Radiol. 2012;85:e837–e844. doi: 10.1259/bjr/13212920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kim HC, Kim TK, Sung KB, Yoon HK, Kim PN, Ha HK, et al. CT during hepatic arteriography and portography: an illustrative review. Radiographics. 2002;22:1041–1051. doi: 10.1148/radiographics.22.5.g02se071041. [DOI] [PubMed] [Google Scholar]
- 29.Park JH, Park YN, Kim MJ, Park MS, Choi JY, Chung YE, et al. Steatotic hepatocellular carcinoma: association of MRI findings to underlying liver disease and clinicopathological characteristics. Liver Int. 2023;43:1332–1344. doi: 10.1111/liv.15589. [DOI] [PubMed] [Google Scholar]
- 30.Taouli B, Johnson RS, Hajdu CH, Oei MT, Merad M, Yee H, et al. Hepatocellular carcinoma: perfusion quantification with dynamic contrast-enhanced MRI. AJR Am J Roentgenol. 2013;201:795–800. doi: 10.2214/AJR.12.9798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Thaiss WM, Kaufmann S, Kloth C, Nikolaou K, Bösmüller H, Horger M. VEGFR-2 expression in HCC, dysplastic and regenerative liver nodules, and correlation with pre-biopsy dynamic contrast enhanced CT. Eur J Radiol. 2016;85:2036–2041. doi: 10.1016/j.ejrad.2016.09.012. [DOI] [PubMed] [Google Scholar]
- 32.Kim CK, Lim JH, Park CK, Choi D, Lim HK, Lee WJ. Neoangiogenesis and sinusoidal capillarization in hepatocellular carcinoma: correlation between dynamic CT and density of tumor microvessels. Radiology. 2005;237:529–534. doi: 10.1148/radiol.2372041634. [DOI] [PubMed] [Google Scholar]
- 33.Tang A, Bashir MR, Corwin MT, Cruite I, Dietrich CF, Do RKG, et al. Evidence supporting LI-RADS major features for CT- and MR imaging-based diagnosis of hepatocellular carcinoma: a systematic review. Radiology. 2018;286:29–48. doi: 10.1148/radiol.2017170554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sakamoto K, Tanaka S, Sato K, Ito E, Nishiyama M, Urakawa H, et al. What is the “washout” of hepatocellular carcinoma as observed on the equilibrium phase CT?: consideration based on the concept of extracellular volume fraction. Jpn J Radiol. 2022;40:1148–1155. doi: 10.1007/s11604-022-01295-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yang HF, Du Y, Ni JX, Zhou XP, Li JD, Zhang Q, et al. Perfusion computed tomography evaluation of angiogenesis in liver cancer. Eur Radiol. 2010;20:1424–1430. doi: 10.1007/s00330-009-1693-y. [DOI] [PubMed] [Google Scholar]
- 36.Pei XQ, Liu LZ, Zheng W, Cai MY, Han F, He JH, et al. Contrast-enhanced ultrasonography of hepatocellular carcinoma: correlation between quantitative parameters and arteries in neoangiogenesis or sinusoidal capillarization. Eur J Radiol. 2012;81:e182–e188. doi: 10.1016/j.ejrad.2011.01.083. [DOI] [PubMed] [Google Scholar]
- 37.Sato N, Marubashi S. What is the optimal surgical treatment for hepatocellular carcinoma beyond the debate between anatomical versus non-anatomical resection? Surg Today. 2022;52:871–880. doi: 10.1007/s00595-021-02352-z. [DOI] [PubMed] [Google Scholar]
- 38.Asayama Y, Yoshimitsu K, Irie H, Nishihara Y, Aishima S, Tajima T, et al. Poorly versus moderately differentiated hepatocellular carcinoma: vascularity assessment by computed tomographic hepatic angiography in correlation with histologically counted number of unpaired arteries. J Comput Assist Tomogr. 2007;31:188–192. doi: 10.1097/01.rct.0000236417.82395.57. [DOI] [PubMed] [Google Scholar]
- 39.Hwang SH, Rhee H. Radiologic features of hepatocellular carcinoma related to prognosis. J Liver Cancer. 2023;23:143–156. doi: 10.17998/jlc.2023.02.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rhee H, An C, Kim HY, Yoo JE, Park YN, Kim MJ. Hepatocellular carcinoma with irregular rim-like arterial phase hyperenhancement: more aggressive pathologic features. Liver Cancer. 2019;8:24–40. doi: 10.1159/000488540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sun HC, Tang ZY, Li XM, Zhou YN, Sun BR, Ma ZC. Microvessel density of hepatocellular carcinoma: its relationship with prognosis. J Cancer Res Clin Oncol. 1999;125:419–426. doi: 10.1007/s004320050296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tanigawa N, Lu C, Mitsui T, Miura S. Quantitation of sinusoid-like vessels in hepatocellular carcinoma: its clinical and prognostic significance. Hepatology. 1997;26:1216–1223. doi: 10.1053/jhep.1997.v26.pm0009362365. [DOI] [PubMed] [Google Scholar]
- 43.Murakami K, Kasajima A, Kawagishi N, Ohuchi N, Sasano H. Microvessel density in hepatocellular carcinoma: prognostic significance and review of the previous published work. Hepatol Res. 2015;45:1185–1194. doi: 10.1111/hepr.12487. [DOI] [PubMed] [Google Scholar]
- 44.Fang JH, Zhou HC, Zhang C, Shang LR, Zhang L, Xu J, et al. A novel vascular pattern promotes metastasis of hepatocellular carcinoma in an epithelial-mesenchymal transition-independent manner. Hepatology. 2015;62:452–465. doi: 10.1002/hep.27760. [DOI] [PubMed] [Google Scholar]
- 45.Zhang P, Ono A, Fujii Y, Hayes CN, Tamura Y, Miura R, et al. The presence of vessels encapsulating tumor clusters is associated with an immunosuppressive tumor microenvironment in hepatocellular carcinoma. Int J Cancer. 2022;151:2278–2290. doi: 10.1002/ijc.34247. [DOI] [PubMed] [Google Scholar]
- 46.Fang JH, Xu L, Shang LR, Pan CZ, Ding J, Tang YQ, et al. Vessels that encapsulate tumor clusters (VETC) pattern is a predictor of sorafenib benefit in patients with hepatocellular carcinoma. Hepatology. 2019;70:824–839. doi: 10.1002/hep.30366. [DOI] [PubMed] [Google Scholar]
- 47.Kawasaki J, Toshima T, Yoshizumi T, Itoh S, Mano Y, Wang H, et al. Prognostic impact of vessels that encapsulate tumor cluster (VETC) in patients who underwent liver transplantation for hepatocellular carcinoma. Ann Surg Oncol. 2021;28:8186–8195. doi: 10.1245/s10434-021-10209-5. [DOI] [PubMed] [Google Scholar]
- 48.Renne SL, Woo HY, Allegra S, Rudini N, Yano H, Donadon M, et al. Vessels encapsulating tumor clusters (VETC) is a powerful predictor of aggressive hepatocellular carcinoma. Hepatology. 2020;71:183–195. doi: 10.1002/hep.30814. [DOI] [PubMed] [Google Scholar]
- 49.Lin WP, Xing KL, Fu JC, Ling YH, Li SH, Yu WS, et al. Development and validation of a model including distinct vascular patterns to estimate survival in hepatocellular carcinoma. JAMA Netw Open. 2021;4:e2125055. doi: 10.1001/jamanetworkopen.2021.25055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liu K, Dennis C, Prince DS, Marsh-Wakefield F, Santhakumar C, Gamble JR, et al. Vessels that encapsulate tumour clusters vascular pattern in hepatocellular carcinoma. JHEP Rep. 2023;5:100792. doi: 10.1016/j.jhepr.2023.100792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med. 2013;19:1450–1464. doi: 10.1038/nm.3391. [DOI] [PubMed] [Google Scholar]
- 52.Sugino T, Yamaguchi T, Ogura G, Saito A, Hashimoto T, Hoshi N, et al. Morphological evidence for an invasion-independent metastasis pathway exists in multiple human cancers. BMC Med. 2004;2:9. doi: 10.1186/1741-7015-2-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Woo HY, Rhee H, Yoo JE, Kim SH, Choi GH, Kim DY, et al. Lung and lymph node metastases from hepatocellular carcinoma: comparison of pathological aspects. Liver Int. 2022;42:199–209. doi: 10.1111/liv.15051. [DOI] [PubMed] [Google Scholar]
- 54.Dennis C, Prince DS, Moayed-Alaei L, Remash D, Carr-Boyd E, Bowen DG, et al. Association between vessels that encapsulate tumour clusters vascular pattern and hepatocellular carcinoma recurrence following liver transplantation. Front Oncol. 2022;12:997093. doi: 10.3389/fonc.2022.997093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kurebayashi Y, Matsuda K, Ueno A, Tsujikawa H, Yamazaki K, Masugi Y, et al. Immunovascular classification of HCC reflects reciprocal interaction between immune and angiogenic tumor microenvironments. Hepatology. 2022;75:1139–1153. doi: 10.1002/hep.32201. [DOI] [PubMed] [Google Scholar]
- 56.Calderaro J, Couchy G, Imbeaud S, Amaddeo G, Letouzé E, Blanc JF, et al. Histological subtypes of hepatocellular carcinoma are related to gene mutations and molecular tumour classification. J Hepatol. 2017;67:727–738. doi: 10.1016/j.jhep.2017.05.014. [DOI] [PubMed] [Google Scholar]
- 57.Calderaro J, Meunier L, Nguyen CT, Boubaya M, Caruso S, Luciani A, et al. ESM1 as a marker of macrotrabecular-massive hepatocellular carcinoma. Clin Cancer Res. 2019;25:5859–5865. doi: 10.1158/1078-0432.CCR-19-0859. [DOI] [PubMed] [Google Scholar]
- 58.Feng Z, Li H, Zhao H, Jiang Y, Liu Q, Chen Q, et al. Preoperative CT for characterization of aggressive macrotrabecular-massive subtype and vessels that encapsulate tumor clusters pattern in hepatocellular carcinoma. Radiology. 2021;300:219–229. doi: 10.1148/radiol.2021203614. [DOI] [PubMed] [Google Scholar]
- 59.Chen FM, Du M, Qi X, Bian L, Wu D, Zhang SL, et al. Nomogram estimating vessels encapsulating tumor clusters in hepatocellular carcinoma from preoperative gadoxetate disodium-enhanced MRI. J Magn Reson Imaging. 2023;57:1893–1905. doi: 10.1002/jmri.28488. [DOI] [PubMed] [Google Scholar]
- 60.Vanderborght B, Lefere S, Vlierberghe HV, Devisscher L. The angiopoietin/TIE2 pathway in hepatocellular carcinoma. Cells. 2020;9:2382. doi: 10.3390/cells9112382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sugimachi K, Tanaka S, Taguchi K, Aishima S, Shimada M, Tsuneyoshi M. Angiopoietin switching regulates angiogenesis and progression of human hepatocellular carcinoma. J Clin Pathol. 2003;56:854–860. doi: 10.1136/jcp.56.11.854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wada H, Nagano H, Yamamoto H, Yang Y, Kondo M, Ota H, et al. Expression pattern of angiogenic factors and prognosis after hepatic resection in hepatocellular carcinoma: importance of angiopoietin-2 and hypoxia-induced factor-1 alpha. Liver Int. 2006;26:414–423. doi: 10.1111/j.1478-3231.2006.01243.x. [DOI] [PubMed] [Google Scholar]
- 63.Villa E, Critelli R, Lei B, Marzocchi G, Cammà C, Giannelli G, et al. Neoangiogenesis-related genes are hallmarks of fast-growing hepatocellular carcinomas and worst survival. Results from a prospective study. Gut. 2016;65:861–869. doi: 10.1136/gutjnl-2014-308483. [DOI] [PubMed] [Google Scholar]
- 64.Llovet JM, Peña CE, Lathia CD, Shan M, Meinhardt G, Bruix J SHARP Investigators Study Group. Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2290–2300. doi: 10.1158/1078-0432.CCR-11-2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Rodríguez-Perálvarez M, Luong TV, Andreana L, Meyer T, Dhillon AP, Burroughs AK. A systematic review of microvascular invasion in hepatocellular carcinoma: diagnostic and prognostic variability. Ann Surg Oncol. 2013;20:325–339. doi: 10.1245/s10434-012-2513-1. [DOI] [PubMed] [Google Scholar]
- 66.Kang I, Jang M, Lee JG, Han DH, Joo DJ, Kim KS, et al. Subclassification of microscopic vascular invasion in hepatocellular carcinoma. Ann Surg. 2021;274:e1170–e1178. doi: 10.1097/SLA.0000000000003781. [DOI] [PubMed] [Google Scholar]
- 67.Hwang YJ, Bae JS, Lee Y, Hur BY, Lee DH, Kim H. Classification of microvascular invasion of hepatocellular carcinoma: correlation with prognosis and magnetic resonance imaging. Clin Mol Hepatol. 2023;29:733–746. doi: 10.3350/cmh.2023.0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Mitsunobu M, Toyosaka A, Oriyama T, Okamoto E, Nakao N. Intrahepatic metastases in hepatocellular carcinoma: the role of the portal vein as an efferent vessel. Clin Exp Metastasis. 1996;14:520–529. doi: 10.1007/BF00115112. [DOI] [PubMed] [Google Scholar]
- 69.Isik B, Gonultas F, Sahin T, Yilmaz S. Microvascular venous invasion in hepatocellular carcinoma: why do recurrences occur? J Gastrointest Cancer. 2020;51:1133–1136. doi: 10.1007/s12029-020-00487-9. [DOI] [PubMed] [Google Scholar]
- 70.WHO Classification of Tumours Editorial Board. WHO classification of tumours of the digestive system. Geneva: World Health Organization; 2019. [Google Scholar]
- 71.Abou-Alfa GK, Pawlik TM, Shindoh J, Vauthey JN. In: AJCC cancer staging manual. 8th ed. Mahul BA, Stephen BE, Frederick LG, David RB, Robert KB, Mary KW, et al., editors. Chicago: American Joint Committee on Cancer; 2017. Liver; pp. 287–293. [Google Scholar]
- 72.Hong SB, Choi SH, Kim SY, Shim JH, Lee SS, Byun JH, et al. MRI features for predicting microvascular invasion of hepatocellular carcinoma: a systematic review and meta-analysis. Liver Cancer. 2021;10:94–106. doi: 10.1159/000513704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lee S, Kang TW, Song KD, Lee MW, Rhim H, Lim HK, et al. Effect of microvascular invasion risk on early recurrence of hepatocellular carcinoma after surgery and radiofrequency ablation. Ann Surg. 2021;273:564–571. doi: 10.1097/SLA.0000000000003268. [DOI] [PubMed] [Google Scholar]
- 74.Cha DI, Kang TW, Jeong WK, Kim JM, Choi GS, Joh JW, et al. Preoperative assessment of microvascular invasion risk using gadoxetate-enhanced MRI for predicting outcomes after liver transplantation for single hepatocellular carcinoma within the Milan criteria. Eur Radiol. 2024;34:498–508. doi: 10.1007/s00330-023-09936-y. [DOI] [PubMed] [Google Scholar]
- 75.Kim AY, Sinn DH, Jeong WK, Kim YK, Kang TW, Ha SY, et al. Hepatobiliary MRI as novel selection criteria in liver transplantation for hepatocellular carcinoma. J Hepatol. 2018;68:1144–1152. doi: 10.1016/j.jhep.2018.01.024. [DOI] [PubMed] [Google Scholar]
- 76.Kim SJ, Kim JM. Prediction models of hepatocellular carcinoma recurrence after liver transplantation: a comprehensive review. Clin Mol Hepatol. 2022;28:739–753. doi: 10.3350/cmh.2022.0060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Thompson SM, Wells ML, Andrews JC, Ehman EC, Menias CO, Hallemeier CL, et al. Venous invasion by hepatic tumors: imaging appearance and implications for management. Abdom Radiol (NY) 2018;43:1947–1967. doi: 10.1007/s00261-017-1298-x. [DOI] [PubMed] [Google Scholar]
- 78.van der Pol CB, Lim CS, Sirlin CB, McGrath TA, Salameh JP, Bashir MR, et al. Accuracy of the liver imaging reporting and data system in computed tomography and magnetic resonance image analysis of hepatocellular carcinoma or overall malignancy—a systematic review. Gastroenterology. 2019;156:976–986. doi: 10.1053/j.gastro.2018.11.020. [DOI] [PubMed] [Google Scholar]
- 79.Reynolds AR, Furlan A, Fetzer DT, Sasatomi E, Borhani AA, Heller MT, et al. Infiltrative hepatocellular carcinoma: what radiologists need to know. Radiographics. 2015;35:371–386. doi: 10.1148/rg.352140114. [DOI] [PubMed] [Google Scholar]
- 80.Catania R, Chupetlovska K, Borhani AA, Maheshwari E, Furlan A. Tumor in vein (LR-TIV) and liver imaging reporting and data system (LI-RADS) v2018: diagnostic features, pitfalls, prognostic and management implications. Abdom Radiol (NY) 2021;46:5723–5734. doi: 10.1007/s00261-021-03270-x. [DOI] [PubMed] [Google Scholar]
- 81.American College of Radiology. Liver Imaging Reporting & Data System (LI-RADS®) [accessed on July 27, 2024]. Available at: https://www.acr.org/Clinical-Resources/Reporting-and-Data-Systems/LI-RADS.
- 82.Bae JS, Lee JM, Jeon SK, Yoo J, Park SJ, Yoon JH, et al. LI-RADS tumor in vein at CT and hepatobiliary MRI. Radiology. 2022;302:107–115. doi: 10.1148/radiol.2021210215. [DOI] [PubMed] [Google Scholar]
- 83.Ichikawa S, Sirlin CB, Cunha GM, Wolfson T, Fowler KJ, Morisaka H, et al. Inter-observer agreement and accuracy of LI-RADS v2018 for differentiating tumor in vein from bland thrombus using gadoxetic acid-enhanced magnetic resonance imaging. Abdom Radiol (NY) 2023;48:2557–2569. doi: 10.1007/s00261-023-03964-4. [DOI] [PubMed] [Google Scholar]
- 84.Bae JS, Lee JM, Yoon JH, Jang S, Chung JW, Lee KB, et al. How to best detect portal vein tumor thrombosis in patients with hepatocellular carcinoma meeting the Milan criteria: gadoxetic acid-enhanced MRI versus contrast-enhanced CT. Liver Cancer. 2020;9:293–307. doi: 10.1159/000505191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kim JH, Lee JM, Yoon JH, Lee DH, Lee KB, Han JK, et al. Portal vein thrombosis in patients with hepatocellular carcinoma: diagnostic accuracy of gadoxetic acid–enhanced MR imaging. Radiology. 2016;279:773–783. doi: 10.1148/radiol.2015150124. [DOI] [PubMed] [Google Scholar]
- 86.Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
- 87.Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet. 2018;391:1163–1173. doi: 10.1016/S0140-6736(18)30207-1. [DOI] [PubMed] [Google Scholar]
- 88.Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;389:56–66. doi: 10.1016/S0140-6736(16)32453-9. [DOI] [PubMed] [Google Scholar]
- 89.Morse MA, Sun W, Kim R, He AR, Abada PB, Mynderse M, et al. The role of angiogenesis in hepatocellular carcinoma. Clin Cancer Res. 2019;25:912–920. doi: 10.1158/1078-0432.CCR-18-1254. [DOI] [PubMed] [Google Scholar]
- 90.Llovet JM, Pinyol R, Kelley RK, El-Khoueiry A, Reeves HL, Wang XW, et al. Molecular pathogenesis and systemic therapies for hepatocellular carcinoma. Nat Cancer. 2022;3:386–401. doi: 10.1038/s43018-022-00357-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Brackenier C, Kinget L, Cappuyns S, Verslype C, Beuselinck B, Dekervel J. Unraveling the synergy between atezolizumab and bevacizumab for the treatment of hepatocellular carcinoma. Cancers (Basel) 2023;15:34. doi: 10.3390/cancers15020348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lee YR. A multidisciplinary approach with immunotherapies for advanced hepatocellular carcinoma. J Liver Cancer. 2023;23:316–329. doi: 10.17998/jlc.2023.09.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kuwano A, Yada M, Miyazaki Y, Tanaka K, Koga Y, Ohishi Y, et al. An imaging feature predicts efficacy of atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. Cancer Diagn Progn. 2023;3:468–474. doi: 10.21873/cdp.10241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.An C, Kim DW, Park YN, Chung YE, Rhee H, Kim MJ. Single hepatocellular carcinoma: preoperative MR imaging to predict early recurrence after curative resection. Radiology. 2015;276:433–443. doi: 10.1148/radiol.15142394. [DOI] [PubMed] [Google Scholar]
- 95.Min JH, Lee MW, Park HS, Lee DH, Park HJ, Lim S, et al. Interobserver variability and diagnostic performance of gadoxetic acid-enhanced MRI for predicting microvascular invasion in hepatocellular carcinoma. Radiology. 2020;297:573–581. doi: 10.1148/radiol.2020201940. [DOI] [PubMed] [Google Scholar]
- 96.Hosoda S, Suda G, Sho T, Ogawa K, Kimura M, Yang Z, et al. Low baseline CXCL9 predicts early progressive disease in unresectable HCC with atezolizumab plus bevacizumab treatment. Liver Cancer. 2022;12:156–170. doi: 10.1159/000527759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 2020;382:1894–1905. doi: 10.1056/NEJMoa1915745. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






