Next Article in Journal
Transcriptomics-Based Drug Repurposing Approach Identifies Novel Drugs against Sorafenib-Resistant Hepatocellular Carcinoma
Next Article in Special Issue
Animal Models: A Useful Tool to Unveil Metabolic Changes in Hepatocellular Carcinoma
Previous Article in Journal
Fusobacterium nucleatum Accelerates the Progression of Colitis-Associated Colorectal Cancer by Promoting EMT
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Hepatocellular Carcinoma: Updates in Pathogenesis, Detection and Treatment

by
Maria L. Martínez-Chantar
1,2,*,
Matias A. Avila
2,3,* and
Shelly C. Lu
4,*
1
CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain
2
National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), 28029 Madrid, Spain
3
Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
4
Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
*
Authors to whom correspondence should be addressed.
Cancers 2020, 12(10), 2729; https://doi.org/10.3390/cancers12102729
Submission received: 15 September 2020 / Accepted: 22 September 2020 / Published: 23 September 2020
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer and the second most common cause of cancer mortality worldwide [1]. The prognosis of HCC patients is very poor. The rates of HCC incidence and mortality are almost equivalent [2] and have increased across most countries over the past three decades [3]. HCC development is closely associated with the presence of chronic liver disease and cirrhosis, albeit the risk factors underlying this condition vary geographically. Hepatitis B virus (HBV) infection and aflatoxin B1 exposure are predominant risk factors in Asia and Africa, while hepatitis C virus (HCV) infection and alcohol consumption are the main risk factors in Europe, the USA and Japan [3,4,5]. Non-alcoholic fatty liver disease (NAFLD) is currently the most prevalent liver disease worldwide, and approximately 60% of biopsied NAFLD patients have non-alcoholic steatohepatitis (NASH) [3]. Importantly, patients with NASH are at high risk of developing HCC even without presenting established cirrhosis [6]. With widespread HBV vaccination and the advent of direct-acting antiviral drugs for HCV infection, NAFLD and associated conditions such as diabetes and obesity are emerging as major global risk factors for HCC. In view of the dismal prognosis of HCC patients, implementing preventive strategies would be an ideal approach to quell the incidence of the disease. Obvious interventions include advocating HBV vaccination in endemic regions, achieving HCV eradication with direct-acting antivirals, promoting healthy nutrition and weight reduction, improving diabetes control, and avoiding excessive alcohol consumption. Still, the implementation of these measures is not always feasible. Chemopreventive strategies have been proposed. However, the identification of chemoprevention targets is not straightforward, and the design and conduction of clinical trials for chemoprevention is resource intensive [3]. HCC screening/surveillance is also essential for early disease detection and the application of effective treatments that reduce disease-associated mortality. For patients with cirrhosis, or with hepatitis B infection without cirrhosis, ultrasonography combined with serum alpha-fetoprotein levels is the currently recommended approach for widespread screening, although computed tomography (CT) and magnetic resonance imaging (MRI) may offer higher sensitivity for small lesions [3,4]. For patients with NAFLD, ultrasonography has decreased diagnostic accuracy, and in these cases, testing for the presence of genetic variants (e.g., I148M of PNPLA3) may inform on HCC risk [7]. The performance of histological biomarkers in combination with clinical data has shown promise for surveillance and early diagnosis [4]. Besides established histoprognostic factors (i.e., tumor differentiation and vascular invasion) the identification of gene signatures with prognostic value has been an area of intense research over the past couple of decades. Nevertheless, mandatory biopsy is not advocated in clinical guidelines, which, together with sampling variability and tumor molecular heterogeneity, have compromised the translation of gene signature assessment into clinical practice [8]. To circumvent these issues, strong efforts are currently being made in the field of liquid biopsy. In this approach, tumor components, including tumor cells, tumor DNA or tumor-derived extracellular vesicles, are analyzed in circulation. It is expected that liquid biopsy may help in early HCC detection, disease prognosis, and treatment response [9,10,11].
From a therapeutic perspective, surgical therapies can be applied in patients with single lesions at an early stage, with well-preserved liver function. However, as many as 70% of these patients develop tumor recurrence after five years [4,8,10]. Therefore, the identification of systemic therapies that may prevent this recurrence, either true recurrence due to dissemination or late recurrence due to de novo tumor formation, is of paramount importance. Locoregional therapies, such as radiofrequency ablation and transarterial therapies, are considered for patients that are not candidates for surgery. Transarterial chemoembolization (TACE), in which cytotoxic agents are injected through tumor-feeding vessels, and selective internal radiation therapy (SIRT) in which microspheres loaded with yttrium-90 are administered, have shown clinical efficacy [4,10]. However, the combination of TACE or SIRT with anti-angiogenic systemic therapies has shown no clinical benefit [8].
When considering the treatment of HCC at more advanced stages, it was clear early on that this tumor is highly resistant to conventional systemic anti-cancer therapies [12]. This realization stimulated the search for more tailored therapies targeting pathways and mechanisms essential for tumor cell survival [12]. Early molecular and cellular studies pointed to interference with growth factor signaling pathways as potential points of pharmacological intervention in anti-HCC therapy [12]. After several failed trials with mono-specific targeted agents, such as epidermal growth factor receptor inhibitors, this approach led to the identification of multikinase inhibitors such as sorafenib, which in carefully designed clinical trials demonstrated efficacy in patients with advanced disease [13]. Sorafenib was followed by other related drugs, such as regorafenib and the even more promiscuous tyrosine kinase inhibitor lenvatinib, which showed improved clinical performance [14]. Experimental and clinical studies suggest the idea that the efficacy of these inhibitors is mediated not only through their interaction with tumor cells, but also with the pro-inflammatory, fibrogenic and pro-angiogenic tumor microenvironment [15]. The unraveling of the genetic features of HCC through the application of high-throughput gene expression analyses and next-generation sequencing (NGS) technologies has provided an increasingly precise molecular portrait. The most recurrent mutations, DNA copy number alterations and associated changes in gene expression contributing to hepatocarcinogenesis, have been described in an increasingly comprehensive manner [16,17,18]. These and other studies allowed for the classification of HCCs into molecular subgroups with associated molecular and clinical phenotypes [13,17]. The most frequent (95% of tumors) and earliest genetic alteration in hepatocarcinogenesis is the overexpression of telomerase reverse transcriptase (TERT) [18]. Unfortunately, pharmacological targeting of TERT has not been successfully achieved yet [17]. More recently, epigenetic alterations are increasingly being recognized as potential driving factors in HCC development, and mutations in epigenetic modifiers can be found in up to 50% of HCCs [19,20]. Importantly, epigenetic mechanisms can be pharmacologically targeted, and the development of the so-called “epidrugs”, already used to treat hematological malignancies, has opened a new avenue for solid tumor therapy, including that of HCC [20,21].
Cancer immunotherapy has also reached the field of HCC treatment. Recent clinical trials have demonstrated promising activity against HCC of immune checkpoint inhibitor (ICI) antibodies targeting cytotoxic T lymphocyte-associated protein-4 (CTLA-4), or anti-programmed cell death protein-1 (PD-1) and its ligand, programmed cell death ligand-1 (PD-L1) [22,23,24]. In spite of being generally well tolerated, the efficacy of ICI-based monotherapy, such as that of multikinase inhibitors, is still suboptimal, and combinatory strategies including ICI and anti-angiogenic agents are actively being pursued [14,23].
Although current HCC therapy is still far from optimal, the promising advances we are witnessing today undoubtedly come from an increased understanding of the fundamental aspects of hepatocarcinogenesis. Better knowledge of tumor cell metabolism [25], HCC epigenetics [26], HCC cancer stem cells [27] and the immunology of HCC, and the hepatic environment [28] will provide new avenues for improved therapeutic intervention. Moreover, further exploration of the role of the gut microbiome and the gut–liver axis in hepatocarcinogenesis will also deliver potential diagnostic tools and therapeutic targets [29]. In this Special Issue “Hepatocellular Carcinoma: Updates in Pathogenesis, Detection and Treatment” of Cancers, we aim to bring together a series of articles providing novel information or insightful reviews of the key aspects of HCC pathogenesis, diagnosis and therapy.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. McGlynn, K.A.; Petrick, J.L.; El-Serag, H.B. Epidemiology of hepatocellular carcinoma. Hepatology 2020. [Google Scholar] [CrossRef]
  2. Golabi, P.; Fazel, S.; Otgonsuren, M.; Sayiner, M.; Locklear, C.T.; Younossi, Z.M. Mortality assessment of patients with hepatocellular carcinoma according to underlying disease and treatment modalities. Medicine 2017, 96. [Google Scholar] [CrossRef]
  3. Fujiwara, N.; Friedman, S.L.; Goossens, N.; Hoshida, Y. Risk factors and prevention of hepatocellular carcinoma in the era of precision medicine. J. Hepatol. 2018, 68, 526–549. [Google Scholar]
  4. Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar]
  5. Avila, M.A.; Dufour, J.F.; Gerbes, A.L.; Zoulim, F.; Bataller, R.; Burra, P.; Cortez-Pinto, H.; Gao, B.; Gilmore, I.; Mathurin, P.; et al. Recent advances in alcohol-related liver disease (ALD): Summary of a Gut round table meeting. Gut 2020, 69, 764–780. [Google Scholar]
  6. Negro, F. Natural history of NASH and HCC. Liver Int. 2020, 40, 72–76. [Google Scholar] [PubMed] [Green Version]
  7. Torres, M.C.P.; Bodini, G.; Furnari, M.; Marabotto, E.; Zentilin, P.; Strazzabosco, M.; Giannini, E.G. Surveillance for hepatocellular carcinoma in patients with non-alcoholic fatty liver disease: Universal or selective? Cancers 2020, 12, 1422. [Google Scholar] [CrossRef]
  8. Gerbes, A.; Zoulim, F.; Tilg, H.; Dufour, J.-F.; Bruix, J.; Paradis, V.; Salem, R.; Peck-Radosavljevic, M.; Galle, P.R.; Greten, T.F.; et al. Gut roundtable meeting paper: Selected recent advances in hepatocellular carcinoma. Gut 2018, 67, 380–388. [Google Scholar] [CrossRef] [PubMed]
  9. Mann, J.; Reeves, H.L.; Feldstein, A.E. Liquid biopsy for liver diseases. Gut 2018, 67, 2204–2212. [Google Scholar] [CrossRef]
  10. Villanueva, A. Hepatocellular carcinoma. N. Engl. J. Med. 2019, 380, 1450–1462. [Google Scholar]
  11. von Felden, J.; Garcia-Lezana, T.; Schulze, K.; Losic, B.; Villanueva, A. Liquid biopsy in the clinical management of hepatocellular carcinoma. Gut 2020. [Google Scholar] [CrossRef] [PubMed]
  12. Avila, M.A.; Berasain, C.; Sangro, B.; Prieto, J. New therapies for hepatocellular carcinoma. Oncogene 2006, 25, 3866–3884. [Google Scholar] [CrossRef] [Green Version]
  13. Llovet, J.M.; Montal, R.; Sia, D.; Finn, R.S. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 2018, 15, 599–616. [Google Scholar] [CrossRef]
  14. Yarchoan, M.; Agarwal, P.; Villanueva, A.; Rao, S.; Dawson, L.A.; Llovet, J.M.; Finn, R.S.; Groopman, J.D.; El-Serag, H.B.; Monga, S.P.; et al. Recent developments and therapeutic strategies against hepatocellular carcinoma. Cancer Res. 2019, 79, 4326–4330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hernandez–Gea, V.; Toffanin, S.; Friedman, S.L.; Llovet, J.M. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology 2013, 144, 512–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ally, A.; Balasundaram, M.; Carlsen, R.; Chuah, E.; Clarke, A.; Dhalla, N.; Holt, R.A.; Jones, S.J.M.; Lee, D.; Ma, Y.; et al. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 2017, 169, 1327–1341.e23. [Google Scholar] [CrossRef]
  17. Dhanasekaran, R.; Nault, J.C.; Roberts, L.R.; Zucman-Rossi, J. Genomic medicine and implications for hepatocellular carcinoma prevention and therapy. Gastroenterology 2019, 156, 492–509. [Google Scholar] [CrossRef]
  18. Rebouissou, S.; Nault, J.C. Advances in molecular classification and precision oncology in hepatocellular carcinoma. J. Hepatol. 2020, 72, 215–229. [Google Scholar] [CrossRef] [Green Version]
  19. Schulze, K.; Imbeaud, S.; Letouzé, E.; Alexandrov, L.B.; Calderaro, J.; Rebouissou, S.; Couchy, G.; Meiller, C.; Shinde, J.; Soysouvanh, F.; et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 2015, 47, 505–511. [Google Scholar] [CrossRef]
  20. Bayo, J.; Fiore, E.J.; Dominguez, L.M.; Real, A.; Malvicini, M.; Rizzo, M.; Atorrasagasti, C.; García, M.G.; Argemi, J.; Martinez, E.D.; et al. A comprehensive study of epigenetic alterations in hepatocellular carcinoma identifies potential therapeutic targets. J. Hepatol. 2019, 71. [Google Scholar] [CrossRef]
  21. Bárcena-Varela, M.; Caruso, S.; Llerena, S.; Álvarez-Sola, G.; Uriarte, I.; Latasa, M.U.; Urtasun, R.; Rebouissou, S.; Alvarez, L.; Jimenez, M.; et al. Dual targeting of histone methyltransferase G9a and DNA-methyltransferase 1 for the treatment of experimental hepatocellular carcinoma. Hepatology 2019, 69, 587–603. [Google Scholar] [CrossRef] [PubMed]
  22. Greten, T.F.; Sangro, B. Targets for immunotherapy of liver cancer. J. Hepatol. 2018, 68, 157–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nakano, S.; Eso, Y.; Okada, H.; Takai, A.; Takahashi, K.; Seno, H. Recent advances in immunotherapy for hepatocellular carcinoma. Cancers 2020, 12, 775. [Google Scholar] [CrossRef] [Green Version]
  24. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef] [PubMed]
  25. Tian, H.; Zhu, X.; Lv, Y.; Jiao, Y.; Wang, G. Glucometabolic reprogramming in the hepatocellular carcinoma microenvironment: Cause and effect. Cancer Manag. Res. 2020, 12, 5957–5974. [Google Scholar] [CrossRef] [PubMed]
  26. Fernández-Barrena, M.; Arechederra, M.; Colyn, L.; Berasain, C.; Avila, M.A. Epigenetics in hepatocellular carcinoma development and therapy: The tip of a big iceberg. J. Hepatol. Rep. 2020, in press. [Google Scholar]
  27. Sia, D.; Villanueva, A.; Friedman, S.L.; Llovet, J.M. Liver cancer cell of origin, molecular class, and effects on patient prognosis. Gastroenterology 2017, 152, 745–761. [Google Scholar] [CrossRef] [Green Version]
  28. Ringelhan, M.; Pfister, D.; O’Connor, T.; Pikarsky, E.; Heikenwalder, M. The immunology of hepatocellular carcinoma review-article. Nat. Immunol. 2018, 19, 222–232. [Google Scholar] [CrossRef]
  29. Schwabe, R.F.; Greten, T.F. Gut microbiome in HCC—Mechanisms, diagnosis and therapy. J. Hepatol. 2020, 72, 230–238. [Google Scholar] [CrossRef] [Green Version]

Share and Cite

MDPI and ACS Style

Martínez-Chantar, M.L.; Avila, M.A.; Lu, S.C. Hepatocellular Carcinoma: Updates in Pathogenesis, Detection and Treatment. Cancers 2020, 12, 2729. https://doi.org/10.3390/cancers12102729

AMA Style

Martínez-Chantar ML, Avila MA, Lu SC. Hepatocellular Carcinoma: Updates in Pathogenesis, Detection and Treatment. Cancers. 2020; 12(10):2729. https://doi.org/10.3390/cancers12102729

Chicago/Turabian Style

Martínez-Chantar, Maria L., Matias A. Avila, and Shelly C. Lu. 2020. "Hepatocellular Carcinoma: Updates in Pathogenesis, Detection and Treatment" Cancers 12, no. 10: 2729. https://doi.org/10.3390/cancers12102729

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop