Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Journal of Gastrointestinal & Digestive System - Hepatocellular carcinoma: a review
ISSN: 2161-069X

Journal of Gastrointestinal & Digestive System
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Review   
  • J Gastrointest Dig Syst 2020, Vol 10(4): 619
  • DOI: 10.4172/2161-069X.1000619

Hepatocellular carcinoma: a review

Stoian Marilena1*, Indrei Lucia2 and Stoica Victor2
1Internal Medicine Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
2Internal Medicine Department, Dr. Ion Cantacuzino Clinical Hospital, Bucharest, Romania
*Corresponding Author: Stoian Marilena, Internal Medicine Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania, Tel: +400733937310, Email: marilenastoian@yahoo.com

Received: 28-Apr-2020 / Accepted Date: 05-Jul-2020 / Published Date: 11-Jul-2020 DOI: 10.4172/2161-069X.1000619

Abstract

Hepatocellular carcinoma is amongst the leading causes of cancer deaths globally, and its incidence is rising not only in Europe, but worldwide, despite the fact that it is preventable, due to the comprehensive understanding of risk factors and underlying liver disease contributing to carcinogenesis. In this paper we present the latest recommendations regarding prevention strategies and screening programs, diagnostic techniques, staging systems and the optimal treatment options for each evolutional stage, with a glimpse into the future directions of the systemic treatment of hepatocellular carcinoma.

Keywords: hepatocellular carcinoma, thermal ablation, transarterial chemoembolization, immune therapy

Introduction

Hepatocellular Carcinoma (HCC) accounts for more than 90% of primary liver cancers, the second most frequent cause of cancer-related death [1]. Although technically any cirrhosis, regardless of its etiology, may be complicated by a malignant tumor development, it is generally accepted that the risk is greater for patients with chronic viral hepatitis, for whom the metabolic syndrome represents an additive risk factor [2,3]. Another frequent risk factor is the alcoholic liver disease and lately the prevalence of HCC amongst the patients with non-alcoholic fatty liver disease (NAFLD) in association with metabolic syndrome has been on the rise, with the possibility of developing HCC even in the absence of cirrhosis [4].

Prevention and Screening

The World Health Organization recommends the vaccination against hepatitis B for all new-borns (even in low-endemic areas) and for the people at high risk of contracting the infection (i.e. IV drug users, health care workers, travelers to endemic areas). Interferon, tenofovir, lamivudine and entecavir have proved useful in the secondary prevention of HCC in patients with chronic hepatitis B, especially associated with cirrhosis [5]. For patients with chronic hepatitis C there was a significant risk reduction of HCC after obtaining a sustained virologic response (SVR) with interferon [6], while controversy persists regarding the role of direct-acting antivirals (DAA). Some authors suggest that the risk of occurrence or recurrence of HCC is higher in patients treated with DAA compared to interferon, with potentially more aggressive tumors in recurrent HCC [7,8] However, a recent meta-analysis did not find any significant difference between interferon and DAA [9], while a retrospective cohort study proved a reduced incidence of HCC in patients who achieved SVR with DAA [10]. There is evidence that coffee reduces not only the incidence of HCC, but also the overall mortality associated with chronic liver disease, although there is no clear dose recommendation [11].

EASL strongly recommends the periodic surveillance of cirrhotic patients (Child-Pugh stages A and B, and only those Child-Pugh stage C patients waiting for a liver transplant), using abdominal ultrasound every six months [2,12,13]. Based on individual risk factors, noncirrhotic patients with NAFLD may be considered candidates for screening.

Diagnosis

In cirrhotic patients with high probability of HCC, the diagnosis can be established only based on non-invasive imagistic criteria: arterial phase hyper-enhancement (APHE) with washout in the portal venous phase on CT or MRI, only for nodules ≥1 cm [14,15]. When CT and MRI are inconclusive, a contrast enhanced ultrasonography (CEUS) showing APHE followed by a mild and late (≥60 seconds) washout is suggestive for HCC [16].

Liver biopsy and histopathological confirmation are mandatory for the HCC diagnosis in non-cirrhotic patients, as well as in cirrhotic patients with inconclusive imaging, remaining the gold standard for defining HCC and the differential diagnosis with other primary liver malignancies (intrahepatic cholangiocarcinoma, combined hepatocellular carcinoma/cholangiocarcinoma) [2]. Immunohistochemistry staining with a three-marker panel (GS, GPC3 and HSP70) can be used to support the diagnosis of early and welldifferentiated HCC [17], while a positive stain for CK19 is associated with a poor prognosis [18]. The potential risks associated with liver biopsy, namely tumor seeding (2.7%), minor hemorrhage (3-4%) or severe hemorrhage requiring transfusion (0.5%), do not affect the course of the disease or the overall survival, are manageable and infrequent, and consequently do not represent a reason to refrain from performing the biopsy [2,19,20].

Histopathology: Precursor lesions

Hepatocellular adenoma (HCA) – although the typical adenomacarcinoma sequence is rarely encountered in HCCs, some HCAs may act as precursor lesions. It appears that a higher risk of malignant transformation is associated with an activating beta-catenin mutation [21]. The evolution of HCA towards HCC is more frequently described in males undergoing androgen treatment or with glycogen storage diseases and in females with oral contraceptive treatment [22]. The differential diagnosis between well-differentiated HCC in noncirrhotic liver and HCA is particularly difficult when the β-catenin-activated hepatocellular adenoma like-tumor occurs in unusual clinical settings (unusual sex or age, i.e. men regardless of age or elderly women, as typical HCA is characteristic for young women) or presents an atypical focal architectural or cytological atypia – these tumors are classified as atypical hepatocellular neoplasm (AHN). There is evidence that in AHN with β-catenin activation there are both morphologic and cytogenetic changes typical for HCCs [23], suggesting that these tumors are actually at least borderline lesions, if not a very highly-differentiated type of HCC, hence the recommendation for tumor resection.

Dysplastic foci – clusters smaller than 1 mm of atypical cells in respect of nuclear size, cytoplasmatic staining or cellular morphology (including the small cell dysplasia typically encountered in cirrhotic livers), but do not meet the criteria for malignancy, lacking the invasive growth [24].

Dysplastic nodules – lesions larger than 1 mm, usually associated with cirrhosis, which can progress to HCC or also regress. Based on their characteristics, dysplastic nodules can be divided into lowgrade and respectively high-grade. Low-grade dysplastic nodules do not present any mitoses, show only minimal atypia and normal or just minimally increased nuclear/cytoplasmatic ratio, with the preservation of the reticulin network and portal tracts. The highgrade dysplastic nodules are characterized by occasional mitoses, peripherally located hyperchromatic nucleus with irregular borders and an increased nuclear/cytoplasmatic ration, basophilic cytoplasm, and occasionally pseudo glandular pattern or unpaired arteries (from which the dysplastic nodules may derive their blood supply) [25,26]. Other cell atypia suggestive changes that can help distinguish highgrade dysplastic nodules from the low-grade ones are focal fatty change (with Mallory-Denk bodies), clear cell change and resistance to iron accumulation.

Macroscopy of HCC

HCCs are typically softer than the surrounding tissue, presenting often a pseudocapsule of fibrous tissue in cirrhotic livers, while in noncirrhotic settings the tumors tend to be unencapsulated. Unifocality and multifocality play an important part in prognosis and the choice of the best therapeutic option. Most of the unifocal HCCs arise from equivocal preexistent nodular lesions, and are usually welldifferentiated at early stages, with the possible multistep evolution to advanced and poorly-differentiated HCCs, especially in patients with chronic HBV or HCV hepatitis [27]. Multifocality is described as tumor nodules which are clearly separated by non-neoplastic liver. The multifocal tumors represent either simultaneously arising independent HCCs known as multicentric HCCs (frequently seen in chronic HBV infections, where the genetic changes induced by the HBV integration patterns are responsible for the independent development of multicentric malignant nodules [28,29]) or intrahepatic metastases from a primary tumor known as satellite tumor nodules. These two distinct possibilities reflect different pathogenic pathways, of utmost importance for treatment and prognosis, as intrahepatic metastases tend to be more often poorly differentiated and more aggressive.

HCCs can be divided into early HCCs (well differentiated, size < 2 cm and vaguely nodular type – poorly defined margins to the surrounding liver) and progressed HCCs (> 2 cm, or size < 2 cm, but moderately differentiated, distinctly nodular type – presents a discernible capsule), according to WHO and international consensus group for hepatocellular carcinoma [30,31]. Macroscopically, progressed HCCs can be nodular, massive or diffuse. The nodular type may comprise a singular encapsulated nodule with or without extracapsular growth adjacent to the nodule or an aggregation of differently sized nodules. The term massive type is generally used to describe a large dominant mass with irregular demarcation with or without smaller satellite nodules, an aspect also encountered in advanced nodular HCC. Diffuse or cirrhotomimetic type is a rare HCC growth pattern characterized by numerous small tumor nodules disseminated throughout the whole liver, closely mimicking the cirrhotic regenerative nodules [25].

Pedunculated HCCs are tumor masses protruding outside the liver with or without a pedicle (some authors use the term protruding HCC if the peduncle is absent). Initially a good prognosis was reported for this type of tumor, although not all the studies have confirmed this outcome [32,33].

Macroscopic findings may be affected by varying grades of portal vein involvement or tumor necrosis, by the accumulation of cytoplasmic glycogen or by the production of bile or fat.

Histopathology of classical HCC

The conventional HCC comprises tumor cells generally resembling hepatocytes, but with cytological atypia and mitotic activity or vascular invasion and stroma consisting of sinusoid-like blood spaces. The endothelial cells of HCC demonstrate changes of “capillarization”, as ultrastructural studies have proven the existence of a basementmembrane- like structure interposed between the trabeculae of tumor cells and the endothelial cells. The angioarchitecture is crucial not only for tumor growth, but also for the imaging techniques. Progressed HCCs are characterized by a prominent neovascularization through unpaired arteries (positive for SMA and CD34 and a smaller amount of elastic fibers than normal intrahepatic arteries), thus appearing hypervascular in imaging, in contrast with the hypovascular appearance of the early HCCs. Typically, there are no portal tracts in the HCC tissue, although they may be present in early HCCs or at the tumor periphery.

HCCs are highly variable from an architectural and cytological point of view, with different architectural patterns and cytological variants occurring frequently at the same patient depending on the different stages of tumor differentiation or in combination. From an immunohistochemmical point of view, around 90% of HCCs are positive for HepPar1 (cytoplasmic positivity with antibodies to carbamoyl phosphate synthetase-1).

The histologic classification of HCC

In addition to the classical or conventional HCC, the WHO admits the existence of 5 morphological subtypes: fibrolamellar HCC (FL-HCC), scirrhous HCC (S-HCC), undifferentiated carcinoma, lymphoepithelioma-like carcinoma and sarcomatoid HCC.

In matters of architectural pattern, HCCs usually present with the following: trabecular or plate-like pattern (most common in well-differentiated or moderate-differentiated HCCs, with trabeculae becoming progressively thicker with de-differentiation; this pattern can be more easily identified using CD34 immunostaining or a reticulin staining), pseudoglandular or acinar pattern (pseudoglandular structures are generally smaller in well-differentiated HCCs than in moderate-differentiated tumors, and their content is frequently PAS+) and compact pattern (usually encountered in poorly differentiated HCCs, where the slit-like sinusoid-like spaces provide the solid appearance of the tumor).

Cytological variants include: pleomorphic cells (typical for poorly differentiated HCCs, without a distinct trabecular pattern, with variably shaped and sized cells lacking cohesiveness, and frequent bizarre multinucleated and giant cells), clear cells (a well-differentiated type of HCC, with cells with a clear cytoplasm due to the accumulation of glycogen, sometimes making it hard to differentiate the HCC from a metastatic renal clear-cell carcinoma, requiring an IHC stain with HepPar1 in order to establish the primary hepatic origin), fatty change (frequent in small, early-stage HCCs, its frequency being inversely proportional with the size of the tumor; probably a consequence of the insufficient blood supply in the initial stages of hepatocarcinogenesis), bile production (yellowish tumor, turning green after fixation with formalin), hyaline or Mallory-Denk bodies (usually irregular intracytoplasmatic eosinophilic and PAS- bodies, positive for antibodies to keratin and ubiquitin, consisting of aggregated intermediary filaments), pale bodies(typical for the fibrolamellar variant of HCC; round-ovoid eosinophilic amorphous material in the cystically dilated endoplasmic reticulum, positive for fibrinogen in IHC) and groundglass inclusions (appearing in HCCs developed in HBsAg-positive patients) [31].

Special types of HCC

Fibrolamellar HCCs – These carcinomas tend to affect younger individuals, with a mean diagnostic age of 25 [34], arising in noncirrhotic livers, with unknown etiology and risk factors, and with a prognosis similar to other HCCs in non-cirrhotic livers, better than classical HCC [35]. The defining histological features of fibrolamellar carcinoma consist of lamellar fibrosis encompassing large polygonal cells with oncocytic cytoplasm and large nuclei [5]. Although in many cases a central scar is present, suggesting a possible connection with focal nodular hyperplasia, these two lesions do not appear to be etiologically related. Focal CK19 and important CK7 expression was reported in this subtype of HCC [36].

Scirrhous HCC – the scirrhous growth pattern describes the prominent fibrosis alongside the sinusoid-like blood spaces, representing more than half of the tumor and encompassing small nests of tumor cells [37]. This histological aspect might be also noticed following transarterial chemoembolization, chemotherapy or radiation, but the post-therapeutic fibrosis needs to be distinguished from the scirrhous histologic subtype. Due to the abundant fibrotic changes of the tumor stroma, scirrhous HCC may closely resemble cholangiocarcinoma, both radiologically and pathologically, but the combination Glypican-3 (GPC-3) + Arginase-1 is useful in the differential diagnosis, having a100% sensitivity for scirrhous HCC [38]. Unlike other HCCs, scirrhous HCC is frequently positive for CK7, while the classical expression of HepPar1 is less common.

Undifferentiated carcinoma – tumors that can be diagnosed as primary carcinomas of the liver on an immunohistochemical basis, but cannot be further classified. They are considered to have a worse outcome compared to classical HCCs.

Lymphoepithelioma-like carcinoma – infrequent type of HCC, characterized by an abundant inflammatory infiltrate consisting mostly of lymphocytes (with the predominance of CD3+ CD4+ lymphocytic subtype), with only a few pleomorphic small tumor cells with focal syncytial growth (and in some cases positive for Epstein-Barr virus). Some authors suggest that this particular aspect might reflect a regression phenomenon [39].

Sarcomatoid HCC – characterized by spindle-shaped, bizarre, anaplastic tumor cells, sometimes hard to distinguish from leiomyosarcoma or fibrosarcoma, especially in the absence of adjacent classical HCC areas. Sarcomatoid changes are more frequently observed in the cases with multiple transarterial chemoembolization or chemotherapy [40].

Clear cell HCC – the defining cytoplasmic clearing is usually the result of glycogen, lipopolysaccharides, mucopolysaccharides or cytoplasmic vesicle accumulation. In order to be classified as a clear cell HCC, the tumor must contain at least 50% cells with clear cytoplasm.

Being a well-differentiated type of HCC, the prognosis of clear cell HCC is better or at least similar when compared to classical HCC [41]. Taking into consideration that there are very few cases of clear cell HCCs arising in hepatitis-free or non-cirrhotic livers [42,43], the presence of a tumor with clear cells within an otherwise normal liver architecture should raise the suspicion of a metastasis from another primary tumor with clear cells, most frequently a clear cell renal or ovarian carcinoma. Usually the HepPar1 expression is useful in distinguishing between a primary liver neoplasia and a metastatic process. However, some ovarian clear cell carcinomas are also positive for HepPar1 staining, making the differential diagnosis more challenging [44,45].

Steatohepatitic HCC – characterized by histological features seen in steatohepatitis representing more than 5% of the tumor, such as Mallory-Denk bodies, inflammation and pericellular fibrosis, ballooning of the hepatocytes and macrovesicular steatosis. A study reported that 63.6% of steatohepatitic HCCs presented with background NAFLD, thus stressing out a possible connection between NAFLD and this particular subtype of HCC [46,47] and consequently the role played by steatohepatitis in hepatocarcinogenesis. However, cases of steatohepatitic HCC have been reported also in the absence of background metabolic disease and non-alcoholic fatty liver disease [48]. The increasing incidence and prevalence of metabolic disease and NAFLD, particularly in western societies, together with the documented observation that HCCs associated with metabolic syndrome or NAFLD can develop even in non-cirrhotic liver, may lead to new recommendations for HCC screening programs [49,50]. The molecular pathway of carcinogenesis of the steatohepatitic HCCs is slightly different: the beta catenin pathway alteration is less frequent compared to classical HCC [51]. Immunohistochemically, steatohepatitic HCCs usually present a more intense staining for amyloid A and C- reactive protein, as well as the conventional staining for HCC (HSP-70, GS, GPC-3). It appears that the prognosis of steatohepatitic HCC is similar to the conventional type of HCC [52].

Staging

After the confirmation of the diagnosis, the next essential step in the management of HCC is to assess the prognosis and indicate the optimal treatment accordingly. There is quite a wide variety of staging systems based on different clinical prognostic factors, with different advantages and disadvantages depending on the statistical model, the study population or the treatment interventional hand [53]. EASL endorses the BCLC (Barcelona-Clinic Liver Cancer) classification (comprising variables regarding the liver function, tumor size and morphological characteristics, as well as the general health performance status), primarily due to the fact that it dynamically links the tumor stage and the treatment option [54].

According to the BCLC classification system, patients with very early stage or stage 0 (preserved liver function i.e. Child-Pugh A without ascites, single nodule less than 2 cm without vascular invasion or satellites, PS 0) can benefit from either ablation or resection and have a mean survival of more than 5 years. In early stage or stage A (preserved liver function, single or 2-3 nodules less than 3 cm, PS 0) the treatment options include resection, transplant and ablation, also with a survival over 5 years. Chemoembolization is best suited for the intermediate stage or stage C (preserved liver function, multinodular and unresectable tumors, PS 0), with a survival of more than 2.5 years. Systemic therapy is reserved for advanced stages i.e. stage C (preserved liver function, but with extrahepatic spread or portal invasion, PS 1-2), with a mean survival a little over 10 months, while in the terminal stage or stage D (end-stage liver function, non-transplantable HCC, PS 3-4) the only option remains the best supportive care, with an estimated survival of approximately 3 months [22,55].

Treatment options

Potentially curative treatment options comprise liver resection, orthotopic liver transplantation and ablation. Although only the liver transplantation addresses both the HCC and the underlying cause and is associated with a benefit in disease-free survival, there was no evidence of its superiority over liver resection in terms of overall survival [56].

In non-cirrhotic patients with HCC liver resection (LR) is the mainstay of treatment, with excellent results and low risks, even in the case of extended resections [57]. However, in the presence of NAFLD, the conditions associated with the metabolic syndrome may negatively influence the outcome [58]. When deciding for a LR in a cirrhotic patient, one should carefully assess the liver function (usually using the Child-Pugh scoring system or MELD score), the presence of clinically relevant portal hypertension associated with the extent of hepatic resection and the estimated remnant hepatic volume respectively (using CT/MRI), and last but not least the patient’s comorbidities and performance status [59]. Technically solitary tumors of any size can benefit from LR, but the outcome varies inversely with the tumor size, provided that a R0 resection can be achieved respecting the liver function-preservation principles [60]. Multinodular HCC does not represent a contraindication for LR, if the nodules fulfill the Milan criteria. Adjuvant therapy is currently not recommended, despite the 50-70% tumor recurrence rate within 5 years following surgery, representing either de novo tumors developed on the remnant cirrhotic liver (more often after 2 years) or true recurrences in the form of intrahepatic metastases (usually appearing earlier, in the first two years following surgery).

Orthotopic liver transplantation (OLT) is a first-line option in selected patients, HCC being the only accepted indication for solid organ transplantation in cancer [61]. The Milan criteria (single lesion ≤5 cm or ≤3 nodules each ≤3cm, without vascular or extrahepatic invasion – extrahepatic metastases and tumor vascular invasion are absolute contraindications for OLT) are currently considered the benchmark for selecting patients and comparing with other criteria [62]. Patients beyond Milan criteria might be considered for OLT in the event of a successful downstaging to within these criteria, or if they meet the more liberal University of California San Francisco (UCSF) criteria (single lesion ≤6.5 cm or ≤3 nodules with the largest tumor ≤4.5 cm and total tumor diameter ≤8 cm, without vascular or extrahepatic invasion) [63]. OLT is severely limited by the low availability of liver allografts, with no clear criteria for prioritizing the allocation of the graft for patients with HCC, although they should include at least the tumor response to treatment, the tumor burden and the waiting time. In order to minimize the risk of tumor progression beyond the Milan criteria and pre-OLT drop-out patients may benefit from “bridging therapies”, such as resection, local ablation or transarterial chemoembolization [64].

Ablation therapies classically comprise radiofrequency ablation (RFA), microwave ablation (MWA) and percutaneous ethanol injection (PEI). Laser ablation, cryoablation and irreversible electroporation have also been proposed for local ablation in HCC. RFA is the gold standard of treatment for BCLC-0 and BCLC-A patients where surgery is not feasible. In terms of efficiency the outcomes of RFA in solitary HCC smaller than 2 cm are at least equal to those of surgery, the location of the tumor being one of the most important factors to consider when deciding between RFA and surgery (due to the “heat sink” effect, RFA is less effective in tumors located less than 1 cm away from a larger vessel i.e. more than 3 mm diameter) [65]. Taking into account the lesser morbidity and invasiveness and also the benefit of destructing the unidentified satellite tumors due to the extension of the ring of necrosis in the peritumoral tissue, RFA could be recommended as firstline treatment in BCLC-0 even when surgery is possible [66]. MWA has proved similar results in terms of local control and overall survival, with a study suggesting the superiority of MWA over RFA in larger tumors [66,67]. Moreover, due to the use of electromagnetic energy for thermal ablation, MWA is less affected by the “heat sink” effect. PEI is usually considered an alternative when thermal ablation is not technically feasible, mostly for tumors smaller than 2 cm, as in larger nodules the tumor necrosis is incomplete, leading to local recurrence in up to 49% of the cases [68,69]. Laser ablation and cryoablation are not inferior to RFA regarding overall survival, but laser ablation requires higher operator skills, while the benefits of cryoablation in local tumor progression are counterbalanced by the risk of “cryoshock” (severe coagulopathy and disseminated intravascular coagulation leading to multi-organ failure) [70-72].

Irreversible electroporation is a new technique that induces cell death by generating pores in the cell lipid bilayer through high-current electrical pulses, presenting the advantage of complete tumor necrosis while preserving the bile ducts in the target area. However, it requires general anesthesia, making it costlier and more demanding than RFA or MWA [73]. In all ablation techniques it is of utmost importance to assess the presence and volume of post-procedural tumor necrosis, using CT, MRI or CEUS.

Transarterial chemoembolization (TACE) is the first treatment option for selected patients with unresectable HCC, namely asymptomatic patients with a good performance status and uni- or pauci-nodular tumors – stage BCLC-B33. TACE can also be used in earlier stages if ablation, OLT or LR are not feasible, or as a bridge to transplant in order to minimize the drop-out risk. Tumor invasion of the main portal branches or impaired portal flow, as well as extrahepatic spread are absolute contraindications for the procedure [74]. The standard of practice still remains the lipiodol-based TACE, although there is evidence the TACE with drug-eluting beads (TACEDEB) is associated with a reduced risk of systemic side effects (mainly cardiac toxicity) of chemotherapy due to the leakage of doxorubicin in the systemic circulation [75]. However, a retrospective study suggests the global hepatic and biliary injuries are more frequent with TACEDEB [76]. Currently there is no clear recommendation regarding the duration and frequency of TACE treatment, but is generally accepted that aggressively repeated TACE procedures induce liver failure and that TACE should not be repeated if there is no substantial tumor necrosis after two sessions or in the event of untreatable progression33. Combining TACE with systemic anti-angiogenic agents is not recommended, but the association of TACE and RFA is beneficial especially for HCCs larger than 3 cm in terms of overall and disease-free survival [77].

The main indication for selective internal radiation therapy (SIRT) is the treatment of locally advanced HCC. In BCLC-B stage SIRT is associated with a better quality of life, improved tumor control and a longer time to progression and less toxicity (SIRT can be safely used even in the event of portal vein thrombosis [78]) when compared to TACE, but this does not translate into a benefit on overall survival [79-81]. Similarly, in BCLC-C stage SIRT has proved a higher response rate than sorafenib, but it does not improve overall survival [82]. In earlier stages SIRT might help reduce the drop-out rate from transplant waiting lists [83] and in borderline resectable HCC SIRT might be associated not only with a better tumor control but also with a significant hypertrophy of the contralateral hepatic lobe [84].

HCC is one of the most chemo-resistant tumors, and systemic chemotherapy is currently not recommended for the treatment of HCC [85], although chemotherapy for HCC in non-cirrhotic patients is underexplored, requiring further research. The standard of care in first-line systemic therapy is sorafenib, a multikinase inhibitor, indicated for patients with good performance status (PS 1-2) and well-preserved liver function (Child-Pugh A) and advanced HCC (BCLC-C) or tumors progressing upon or unsuitable for locoregional therapies [33,85]. Patients without extrahepatic spread or those infected with HCV tend to benefit more from the therapy with sorafenib [86]. Sorafenib should be administered until radiologic progression of the lesions, when second-line therapy with regorafenib is recommended. Levantinib has demonstrated non-inferiority to sorafenib as first-line therapy in terms of overall survival for patients with advanced HCC (excluding a tumor burden >50% of liver volume or main portal vein invasion), PS 0-1 and Child-Pugh A, and was associated with higher response rates, longer time to progression and improved disease-free survival [87]. Second-line systemic therapies include regorafenib (approved for patients who tolerated sorafenib but experienced radiologic progression on sorafenib, with PS 0-1 and Child-Pugh A) [88], cabozantinib (as monotherapy for advanced or unresectable HCC, for patients previously treated with sorafenib) [89] and ramucirumab (for patients with advanced or unresectable HCC, baseline AFP ≥400 ng/ml and previously treated with sorafenib) [90]. Immune therapies with nivolumab, pembrolizumab and the dual therapy nivolumab+ipilimumab have recently received FDA approval as second-line therapy in advanced HCC, but EASL considers that the current data is insufficient for a clear recommendation.

Future directions in systemic therapies are based on identifying new pathways involved in the carcinogenesis of HCC that could become therapeutic targets. The higher prevalence of HCC in men has led to studies exploring the androgen receptor expression in HCC and a phase II study with enzalutamide [91]. Glypican 3 is a membrane protein involved in cell proliferation and a possible target for immunotherapy in HCC, with an ongoing research on manipulating the T-cell receptor to recognize glypican 3 [92-94]. Another interesting immunotherapy concept consists of the use of oncolytic viruses: T-VEC (a modified herpes virus) and Pexa-vec (a modified vaccinia virus), that at sufficient infectious doses are capable of inducing the autolysis of the tumor [95,96]. Sapanisertib, a strong mTOR inhibitor, has proved in vitro efficacy in HCC lines resistant to sorafenib [97], just like galunisertib [98], inhibitor of TGF-β signaling pathway, making these two drugs candidates for clinical trials.

Conclusion

In most of the cases HCC develops in the context of a welldefined underlying liver disease, thus being preventable. However, the incidence of HCC has been rising in the past years in the most European countries, pointing out the lack of awareness of the general population regarding liver diseases in general and HCC in particular. Due to the inappropriate surveillance for cancer and neglect of liver disease many patients with HCC are diagnosed at a more advanced stage, when curative treatment is no longer an option. These epidemiological observations underline the imperative need for public health policies addressed to preventing, detecting and treating chronic liver disease and for appropriate screening programs allowing the diagnosis of HCC at an earlier stage, suitable for curative treatment. Other major challenges in the management of HCC are represented by the need to optimally describe the sequencing of systemic therapy and the development of third-line therapies in advanced HCC. Ongoing research will probably provide new drugs targeting a wide variety of presumed pathogenic mechanisms involved in HCC carcinogenesis, but managing the irreversible cirrhosis and the hepatotoxic side effects will definitely remain crucial issues to address for each and every new therapeutic agent.

References

  1. Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, et al. (2017) The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level. JAMA Oncol 3:1683–1691.
  2. EASL (2018) Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol 69:182-236.
  3. Chen C, Yang H, Yang W, Liu C, Chen P, et al (2008) Metabolic factors and risk of hepatocellular carcinoma by chronic hepatitis B/C infection:a follow-up study in Taiwan. Gastroenterology 135:111–121.
  4. Degasperi E (2016) Colombo M. Distinctive features of hepatocellular carcinoma in non-alcoholic fatty liver disease. Lancet Gastroenterol Hepatol 1:156–164.
  5. EASL 2017 (2017) Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol 2017: 370–398.
  6. van der Meer AJ, Veldt BJ, Feld JJ, Wedemeyer H, Dufour J-F, et al. (2012) Association between sustained virological response and all-cause mortality among patients with chronic hepatitis C and advanced hepatic fibrosis. JAMA 308:2584.
  7. Reig M, Marino Z, Perello C, Inarrairaegui M, Ribeiro A, et al. ( 2016) Unexpected high rate of early tumor recurrence in patients with HCVrelated HCC undergoing interferon-free therapy. J Hepatol 65:719–726.
  8. Conti F, Buonfiglioli F, Scuteri A, Crespi C, Bolondi L, et al. (2016) Early occurrence and recurrence of hepatocellular carcinoma in HCVrelated cirrhosis treated with direct-acting antivirals. J Hepatol 65:727–733.
  9. Waziry R, Hajarizadeh B, Grebely J, Amin J, Law M, et al. (2017) Hepatocellular carcinoma risk following direct-acting antiviral HCV therapy: A systematic review, meta-analyses, and meta-regression. J Hepatol 67:1204–1212.
  10. Kanwal F, Kramer J, Asch SM, Chayanupatkul M, Cao Y, et al. (2017) Risk of hepatocellular cancer in HCV patients treated with direct-acting antiviral agents. Gastroenterology 153:996–1005.
  11. Setiawan VW, Wilkens LR, Lu SC, Hernandez BY, Le Marchand L, et al. (2015) Association of coffee intake with reduced incidence of liver cancer and death from chronic liver disease in the US multiethnic cohort. Gastroenterology 148:118–25.
  12. Thompson Coon J, Rogers G, Hewson P, Wright D, Anderson R, et al. (2008) Surveillance of cirrhosis for hepatocellular carcinoma: a costutility analysis. Br J Cancer 98:1166–1175.
  13. Trinchet JC, Chaffaut C, Bourcier V, Degos F, Henrion J, et al. (2011) Ultrasonographic surveillance of hepatocellular carcinoma in cirrhosis: a randomized trial comparing 3- and 6-month periodicities. Hepatology 54:1987–1997.
  14. Matsui O, Kobayashi S, Sanada J, Kouda W, Ryu Y, et al. (2011) Hepatocelluar nodules in liver cirrhosis: hemodynamic evaluation (angiography-assisted CT) with special reference to multi-step hepatocarcinogenesis. Abdom Imaging 36:264–272.
  15. Forner A, Vilana R, Ayuso C, Bianchi L, Sole M, et al. (2008) Diagnosis of hepatic nodules 20 mm or smaller in cirrhosis: Prospective validation of the noninvasive diagnostic criteria for hepatocellular carcinoma. Hepatology 47:97–104.
  16. Furlan A, Marin D, Cabassa P, Taibbi A, Brunelli E, et al. (2012) Enhancement pattern of small hepatocellular carcinoma (HCC) at contrast-enhanced US (CEUS), MDCT, and MRI: Intermodality agreement and comparison of diagnostic sensitivity between 2005 and 2010 American Association for the Study of Liver Diseases (AASLD). Eur J Radiol 81:2099–2105.
  17. Tremosini S, Forner A, Boix L, Vilana R, Bianchi L, et al. (2012) Prospective validation of an immunohistochemical panel (glypican 3, heat shock protein 70 and glutamine synthetase) in liver biopsies for diagnosis of very early hepatocellular carcinoma. Gut 61:1481–1487.
  18. Durnez A, Verslype C, Nevens F, Fevery J, Aerts R, et al. (2006) The clinicopathological and prognostic relevance of cytokeratin 7 and 19 expression in hepatocellular carcinoma. A possible progenitor cell origin. Histopathology 49:138–151.
  19. Silva MA, Hegab B, Hyde C, Guo B, Buckels JAC, Mirza DF (2008) Needle track seeding following biopsy of liver lesions in the diagnosis of hepatocellular cancer: a systematic review and meta-analysis. Gut 57:1592–1596.
  20. Fuks D, Cauchy F, Fusco G, Paradis V, Durand F, Belghiti J (2014) Preoperative tumour biopsy does not affect the oncologic course of patients with transplantable HCC. J Hepatol 61:589–593.
  21. Zucman-Rossi J, Jeannot E, Nhieu JT, Scoazec JY, Guettier C, et al. (2006) Genotype-phenotype correlation in hepatocellular adenoma: new classification and relationship with HCC. Hepatology 43:515–524.
  22. Chang CY, Hernandez-Prera JC, Roayaie S, Schwartz M, Thung SN (2013) Changing epidemiology of hepatocellular adenoma in the United States: review of the literature. Int J Hepatol 2013:604860.
  23. Evason KJ, Grenert JP, Ferrell LD, Kakar S (2013) Atypical hepatocellular adenoma-like neoplasms with β-catenin activation show cytogenetic alterations similar to well-differentiated hepatocellular carcinomas. Hum Pathol 44:750–758
  24. International Working Party (1995) Terminology of nodular hepatocellular lesions. Hepatology 22:983–993
  25. Kojiro M (2009) Pathology of Hepatocellular Carcinoma. Hoboken: Wiley P.No: 1–184.
  26. Roncalli M (2004) Hepatocellular nodules in cirrhosis: focus on diagnostic criteria on liver biopsy: A Western experience. Liver Transpl 10:S9–S15.
  27. Oikawa T, Ojima H, Yamasaki S, Takayama T, Hirohashi S, et al. (2005) Multistep and multicentric development of hepatocellular carcinoma: histological analysis of 980 resected nodules. J Hepatol 42:225–229.
  28. Ng IO, Guan XY, Poon RT, Fan ST, Lee JM (2003) Determination of the molecular relationship between multiple tumour nodules in hepatocellular carcinoma differentiates multicentric origin from intrahepatic metastasis. J Pathol 199:345–353.
  29. Sakamoto M, Hirohashi S, Tsuda H, Shimosato Y, Makuuchi M, et al. (1989) Multicentric independent development of hepatocellular carcinoma revealed by analysis of hepatitis B virus integration pattern. Am J Surg Pathol 13:1064–1067
  30. The International Agency for Research on Cancer (2010) WHO Classification of Tumours of the Digestive System (IARC WHO Classification of Tumours). 4th ed. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, editors. Publisher: World Health Organization pp. 1–418.
  31. International Consensus Group for Hepatocellular NeoplasiaThe International Consensus Group for Hepatocellular Neoplasia (2009) Pathologic diagnosis of early hepatocellular carcinoma: a report of the international consensus group for hepatocellular neoplasia. Hepatology 49:658–664.
  32. Yeh CN, Lee WC, Jeng LB, Chen MF (2002) Pedunculated hepatocellular carcinoma: clinicopathologic study of 18 surgically resected cases. World J Surg 26(9):1133‐1138.
  33. Horie Y, Shigoku A, Tanaka H, Tomie Y, Maeda N, et al. (1999) Prognosis for pedunculated hepatocellular carcinoma. Oncology 57(1):23‐28.
  34. Eggert T, McGlynn KA, Duffy A, Manns MP, Greten TF, et al. (2013) Fibrolamellar hepatocellular carcinoma in the USA, 2000-2010: A detailed report on frequency, treatment and outcome based on the Surveillance, Epidemiology, and End Results database. United European Gastroenterol J 1:351–357.
  35. Kakar S, Burgart LJ, Batts KP, Garcia J, Jain D, et al. (2005) Clinicopathologic features and survival in fibrolamellar carcinoma: Comparison with conventional hepatocellular carcinoma with and without cirrhosis. Mod Pathol 18:1417–1423.
  36. Van Eyken P, Sciot R, Brock P, Casteels-Van Daele M, Ramaekers FC, et al. (1990) Abundant expression of cytokeratin 7 in fibrolamellar carcinoma of the liver. Histopathology 17:101–107
  37. Torbenson M, Zen Y, Yeh MM.( 2018) Washington (DC): American Registry of Pathology; Tumors of the liver, AFIP Atlas of tumor pathology series 4; pp. 39–112.
  38. Krings G, Ramachandran R, Jain D, Wu TT, Yeh MM, et al. (2013) Immunohistochemical pitfalls and the importance of glypican 3 and arginase in the diagnosis of scirrhous hepatocellular carcinoma. Mod Pathol 26:782–791.
  39. Park HS, Jang KY, Kim YK, Cho BH, Moon WS (2009) Hepatocellular carcinoma with massive lymphoid infiltration: a regressing phenomenon? Pathol Res Pract 205:648–652.
  40. Nishi H, Taguchi K, Asayama Y, Aishima S, Sugimachi K, et al. (2003) Sarcomatous hepatocellular carcinoma: a special reference to ordinary hepatocellular carcinoma. J Gastroenterol Hepatol 18:415–423.
  41. Ji SP, Li Q, Dong H (2010) Therapy and prognostic features of primary clear cell carcinoma of the liver. World J Gastroenterol 16:764–769.
  42. Clayton EF, Furth EE, Ziober A, Xu T, Yao Y, et al. (2012) case of primary clear cell hepatocellular carcinoma in a non-cirrhotic liver: An immunohistochemical and ultrastructural study. Rare Tumors 4:e29.
  43. Takahashi A, Saito H, Kanno Y, Abe K, Yokokawa J, et al. (2008) Case of clear-cell hepatocellular carcinoma that developed in the normal liver of a middle-aged woman. World J Gastroenterol 14:129–131.
  44. Murakata LA, Ishak KG, Nzeako UC (2000) Clear cell carcinoma of the liver: A comparative immunohistochemical study with renal clear cell carcinoma. Mod Pathol 13:874–881.
  45. Fan Z, van de Rijn M, Montgomery K, Rouse RV (2003) Hep par 1 antibody stain for the differential diagnosis of hepatocellular carcinoma: 676 tumors tested using tissue microarrays and conventional tissue sections. Mod Pathol 16:137–144.
  46. Salomao M, Yu WM, Brown RS, Jr, Emond JC, Lefkowitch JH (2010) Steatohepatitic hepatocellular carcinoma (SH-HCC): A distinctive histological variant of HCC in hepatitis C virus-related cirrhosis with associated NAFLD/NASH. Am J Surg Pathol 34:1630–1636.
  47. Salomao M, Remotti H, Vaughan R, Siegel AB, Lefkowitch JH, et al. (2012) The steatohepatitic variant of hepatocellular carcinoma and its association with underlying steatohepatitis. Hum Pathol 43:737–746.
  48. Yeh MM, Liu Y, Torbenson M (2015) Steatohepatitic variant of hepatocellular carcinoma in the absence of metabolic syndrome or background steatosis: A clinical, pathological, and genetic study. Hum Pathol 46:1769–1775.
  49. Paradis V, Zalinski S, Chelbi E, Guedj N, Degos F, et al. (2009) Hepatocellular carcinomas in patients with metabolic syndrome often develop without significant liver fibrosis: A pathological analysis. Hepatology 49:851–859.
  50. Baffy G, Brunt EM, Caldwell SH (2012) Hepatocellular carcinoma in non-alcoholic fatty liver disease: An emerging menace. J Hepatol 56:1384–1391.
  51. Ando S, Shibahara J, Hayashi A, Fukayama M (2015) β-catenin alteration is rare in hepatocellular carcinoma with steatohepatitic features: Immunohistochemical and mutational study. Virchows Arch 467:535–542.
  52. Taniai M, Hashimoto E, Tobari M, Kodama K, Tokushige K, et al. (2018) Clinicopathological investigation of steatohepatitic hepatocellular carcinoma: A multicenter study using immunohistochemical analysis of adenoma-related markers. Hepatol Res 48:947–955.
  53. Hsu CY, Hsia CY, Huang YH, et al. (2010) Selecting an optimal staging system for hepatocellular carcinoma: comparison of 5 currently used prognostic models. Cancer 116:3006–14.
  54. Llovet JM, Bru C, Bruix J (1999) Prognosis of hepatocellular carcinoma: the BCLC staging classification. Semin Liver Dis 19:329–338.
  55. Forner A, Reig M, Bruix J (2018) Hepatocellular carcinoma. Lancet 391:1301-1314.
  56. Rahbari NN, Mehrabi A, Mollberg NM, Müller SA, Koch M, et al. (2011) Hepatocellular carcinoma: current management and perspectives for the future. Ann Surg 253: 453–469.
  57. Lang H, Sotiropoulos GC, Domland M, Fruhauf NR, Paul A, et al. (2005) Liver resection for hepatocellular carcinoma in non-cirrhotic liver without underlying viral hepatitis. Br J Surg 92:198–202.
  58. Cauchy F, Zalinski S, Dokmak S, Fuks D, Farges O, et al. (2013) Surgical treatment of hepatocellular carcinoma associated with the metabolic syndrome. Br J Surg 100:113–121.
  59. Citterio D, Facciorusso A, Sposito C, Rota R, Bhoori S, et al. (2016) Hierarchic interaction of factors associated with liver decompensation after resection for hepatocellular carcinoma. JAMA Surg 151:846.
  60. Vitale A, Burra P, Frigo AC, Trevisani F, Farinati F, et al. (2015) Survival benefit of liver resection for patients with hepatocellular carcinoma across different Barcelona Clinic Liver Cancer stages: a multicentre study. J Hepatol 62:617–624.
  61. Mazzaferro V, Battiston C, Sposito C (2018) Pro (With Caution): Extended oncologic indications in liver transplantation. Liver Transplant 24:98–103
  62. Mazzaferro V, Bhoori S, Sposito C, Bongini M, Langer M, et al. (2011) Milan criteria in liver transplantation for hepatocellular carcinoma: An evidence-based analysis of 15 years of experience. Liver Transpl 17: S44–S57.
  63. Yao FY, Xiao L, Bass NM, Kerlan R, Ascher N L, Roberts J P, et al. (2007) Liver transplantation for hepatocellular carcinoma: validation of the UCSF-expanded criteria based on preoperative imaging. Am J Transplant 7: 2587–2596.
  64. Vogel A, Cervantes A, Chau I, Daniele B, Llovet J M, et al. (2018) Hepatocellular carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 29(Suppl 4): iv238-iv255.
  65. Cucchetti A, Piscaglia F, Cescon M, Colecchia A, Ercolani G, et al. (2013) Cost-effectiveness of hepatic resection vs. percutaneous radiofrequency ablation for early hepatocellular carcinoma. J Hepatol 59:300–307.
  66. Lee DH, Lee JM, Lee JY, Kim SH, Yoon JH, et al. (2014) Radiofrequency ablation of hepatocellular carcinoma as first-line treatment: long-term results and prognostic factors in 162 patients with cirrhosis. Radiology 270:900–909.
  67. Poulou LS, Botsa E, Thanou I, Ziakas PD, Thanos L (2015) Percutaneous microwave ablation vs. radiofrequency ablation in the treatment of hepatocellular carcinoma. World J Hepatol 7:1054–1063.
  68. Facciorusso A, Di Maso M, Muscatiello N (2016) Microwave ablation vs. radiofrequency ablation for the treatment of hepatocellular carcinoma: A systematic review and meta-analysis. Int J Hyperthermia 32:339–344.
  69. Pompili M, De Matthaeis N, Saviano A, De Sio I, Francica G, et al. (2015) Single hepatocellular carcinoma smaller than 2 cm: are ethanol injection and radiofrequency ablation equally effective? Anticancer Res 35:325–332.
  70. Di Costanzo GG, Tortora R, D’Adamo G, De Luca M, Lampasi F, et al. (2015) Radiofrequency ablation vs. laser ablation for the treatment of small hepatocellular carcinoma in cirrhosis: a randomized trial. J Gastroenterol Hepatol 30:559–565.
  71. Francica G, Petrolati A, Di Stasio E, Pacella S, Stasi R, et al. (2012) Effectiveness, safety, and local progression after percutaneous laser ablation for hepatocellular carcinoma nodules up to 4 cm are not affected by tumor location. AJR Am J Roentgenol 199:1393–1401.
  72. Wang C, Wang H, Yang W, Hu K, Xie H, et al. (2015) Multicenter randomized controlled trial of percutaneous cryoablation vs. radiofrequency ablation in hepatocellular carcinoma. Hepatology 61: 1579–1590.
  73. Cheng RG, Bhattacharya R, Yeh MM, Padia SA (2015) Irreversible electroporation can effectively ablate hepatocellular carcinoma to complete pathologic necrosis. J Vasc Interv Radiol 26:1184–1188.
  74. Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, et al. (2002) Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology 35:1164–1171.
  75. Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, et al. (2010) Prospective randomized study of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of the PRECISION V study. Cardiovasc Intervent Radiol 33: 41–52.
  76. Monier A, Guiu B, Duran R, Aho S, Bize P, et al. (2017) Liver and biliary damages following transarterial chemoembolization of hepatocellular carcinoma: comparison between drug-eluting beads and lipiodol emulsion. Eur Radiol 27:1431–1439.
  77. Wang X, Hu Y, Ren M, Lu X, Lu G, et al. (2016) Efficacy and safety of radiofrequency ablation combined with transcatheter arterial chemoembolization for hepatocellular carcinomas compared with radiofrequency ablation alone: a time-to-event meta-analysis. Korean J Radiol 17:93.
  78. Kulik LM, Carr BI, Mulcahy MF, Lewandowski RJ, Atassi B, et al. (2008) Safety and efficacy of 90Y radiotherapy for hepatocellular carcinoma with and without portal vein thrombosis. Hepatology 47:71–81.
  79. Salem R, Lewandowski RJ, Mulcahy MF, Riaz A, Ryu RK, et al. (2010) Radioembolization for hepatocellular carcinoma using Yttrium-90 microspheres: a comprehensive report of long-term outcomes. Gastroenterology 138:52–64.
  80. Sangro B, Carpanese L, Cianni R, Golfieri R, Gasparini D, et al. (2011) Survival after yttrium-90 resin microsphere radioembolization of hepatocellular carcinoma across Barcelona clinic liver cancer stages: a European evaluation. Hepatology 54:868–878.
  81. Hilgard P, Hamami M, Fouly El A, Scherag A, Muller S, et al. (2010) Radioembolization with yttrium-90 glass microspheres in hepatocellular carcinoma: European experience on safety and long-term survival. Hepatology 52:1741–1749.
  82. Vilgrain V, Pereira H, Assenat E, Guiu B, Ilonca AD, et al. (2017) Efficacy and safety of selective internal radiotherapy with yttrium-90 resin microspheres compared with sorafenib in locally advanced and inoperable hepatocellular carcinoma (SARAH): an open-label randomized controlled phase 3 trial. Lancet Oncol 18:1624–36.
  83. Salem R, Gordon AC, Mouli S, Hickey R, Kallini J, et al. (2016) Y90 radioembolization significantly prolongs time to progression compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology 151:1155–1163.
  84. Garlipp B, de Baere T, Damm R, Irmscher R, van Buskirk M, et al. (2014) Left-liver hypertrophy after therapeutic right-liver radioembolization is substantial but less than after portal vein embolization. Hepatology 59:1864–1873.
  85. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, et al. (2008) Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 359: 378–390.
  86. Bruix J, Cheng AL, Meinhardt G, Nakajima K, De Sanctis Y, et al. (2017) Prognostic factors and predictors of sorafenib benefit in patients with hepatocellular carcinoma: analysis of two phase III studies. J Hepatol 67: 999–1008.
  87. Kudo M, Finn RS, Qin S, Han K-H, Ikeda K, et al. (2018) Lenvatinib vs. sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet 391: 1163-1173.
  88. Bruix J, Qin S, Merle P, Granito A, Huang Y-H, et al. (2017) Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, doubleblind, placebo-controlled, phase 3 trial. Lancet 7;389: 56–66.
  89. Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry A, Rimassa L, et al. (2018) Cabozantinib (C) vs. placebo (P) in patients (pts) with advanced hepatocellular carcinoma (HCC) who have received prior sorafenib: Results from the randomized phase III CELESTIAL trial. J Clin Oncol 36: 4019-4019.
  90. Zhu AX, Kang Y-K, Yen C-J, Finn RS, Galle PR, et al. (2018) REACH-2: a randomized, doubleblind, placebo-controlled phase 3 study of ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma (HCC) and elevated baseline alpha-fetoprotein (AFP) following first-line sorafenib. J Clin Oncol 36: 4003–4003.
  91. Zhang H, Li XX, Yang Y, Zhang Y, Wang HY, et al. (2018) Significance and mechanism of androgen receptor overexpression and androgen receptor/mechanistic target of rapamycin cross-talk in hepatocellular carcinoma. Hepatology 67:2271–86.
  92. Gao H, Li K, Tu H, Pan X, Jiang H, et al. (2014) Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res20:6418–28.
  93. Dargel C, Bassani-Sternberg M, Hasreiter J, Zani F, Bockmann JH, et al. (2015) T cells engineered to express a T-cell receptor specific for glypican-3 to recognize and kill hepatoma cells in vitro and in mice. Gastroenterology149:1042–52.
  94. Bi Y, Jiang H, Wang P, Song B, Wang H, et al. (2017) Treatment of hepatocellular carcinoma with a GPC3-targeted bispecific T cell engager. Oncotarget.8:52866–76.
  95. Heo J, Reid T, Ruo L, Rose S, Bloomston M, et al. (2013) Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med 19:329–36.
  96. Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, et al. (2015) Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 33:2780–8.
  97. Badawi M, Kim J, Dauki A, Sutaria D, Motiwala T, et al. (2018) CD44 positive and sorafenib insensitive hepatocellular carcinomas respond to the ATP-competitive mTOR inhibitor INK128. Oncotarget 9:26032–45.
  98. Serova M, Tijeras-Raballand A, Dos Santos C, Albuquerque M, Paradis V, et al. (2015) Effects of TGF-beta signalling inhibition with galunisertib (LY2157299) in hepatocellular carcinoma models and in ex vivo whole tumor tissue samples from patients. Oncotarget 6:21614–27.

Citation: Marilena S, Lucia I, Victor S (2020) Hepatocellular Carcinoma: A Review. J Gastrointest Dig Syst 10: 619. DOI: 10.4172/2161-069X.1000619

Copyright: © 2020 Marilena S, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top