Open access peer-reviewed chapter

Non-alcoholic Fatty Liver Disease Associated Hepatocellular Carcinoma

Written By

Kai Sun, Alan Hodges and Maen Abdelrahim

Submitted: 26 June 2022 Reviewed: 27 July 2022 Published: 13 April 2023

DOI: 10.5772/intechopen.106816

From the Edited Volume

Liver Cancer - Genesis, Progression and Metastasis

Edited by Mark Feitelson and Alla Arzumanyan

Chapter metrics overview

65 Chapter Downloads

View Full Metrics

Abstract

Non-alcoholic fatty liver disease (NAFLD) is a spectrum of diseases ranging from non-alcoholic fatty liver and non-alcoholic steatohepatitis to its more severe forms such as liver fibrosis and cirrhosis. The incidence of hepatocellular carcinoma (HCC) increases as NAFLD progresses to the more severe forms. As prevalence of obesity and metabolic syndrome rising in North America, NAFLD associated HCC is becoming the leading cause of HCC. Different from other causes of HCC, altered metabolic state and its impact on immune response play an important role in the pathogenesis of NAFLD associated HCC. Currently, immune checkpoint inhibitors and combination therapy are first-line treatments of advanced HCC regardless of etiologies. Given the rising incidence of NAFLD associated HCC and its unique pathogenesis, future clinical trials should assess whether HCC etiology—NAFLD in particular—influence the safety and efficacy of a given treatment.

Keywords

  • hepatocellular carcinoma
  • non-alcoholic fatty liver disease
  • non-alcoholic steatohepatitis
  • liver cirrhosis
  • tyrosine kinase inhibitor
  • immunotherapy

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is defined as the presence of >= 5% hepatic fat accumulation of the liver in the absence of other causes of fatty liver disease. NAFLD is a spectrum of diseases. In its mildest form- non-alcoholic fatty liver (NAFL, also known as simple hepatic steatosis)- fat accumulation is seen in the liver but without significant inflammation or hepatocellular injury. The next stage of NAFLD is non-alcoholic steatohepatitis (NASH) which is characterized by histological lobular inflammation and hepatocyte ballooning. The condition then progresses to liver fibrosis and accumulation of fibrosis leads to liver cirrhosis [1]. In studies [2, 3, 4, 5] that examined paired liver biopsies in patients with baseline NAFLD, up to 20–40% patients with NAFL can progress to fibrosis over an average follow-up between 2.2 and 13.8 years. In a meta-analysis including 411 patients with NAFLD, 35.8%, 32.5%, 16.7%, 9.3%, and 5.7% patients exhibited stage 0,1,2,3,4 fibrosis, respectively with an average fibrosis progression of 0.07 stages per year [6]. The rate of progression is twice as high in patients with NASH and a subgroup of both NASH and NAFL patients may rapidly progress from no fibrosis to advanced fibrosis over an average of six years [3, 7].

NAFLD is considered the hepatic manifestation of metabolic syndrome and many other manifestations of metabolic syndrome including obesity and type 2 diabetes mellitus (T2DM) are independent risk factors of NAFLD. As metabolic syndrome becomes epidemic, the predicted prevalence of obesity will reach close to 50% by year 2030 [8], and the projected NAFLD prevalence among the adult population (aged ≥15 years) will rise to 33.5% [9]. NAFLD-associated hepatocellular carcinoma (HCC) cases are expected to increase by 146% from 10,100 to 24,900 during 2015–2030 and become the leading cause of HCC.

This chapter summaries the current knowledge on the diagnosis, epidemiology, pathogenesis, and treatment of NAFLD associated HCC. It highlights the unique pathogenesis of NALFD associated HCC and discusses challenges we face with current treatment.

Advertisement

2. Definition and diagnosis of NAFLD

Liver biopsy remains the gold standard of diagnosing NAFL, NASH, liver fibrosis and cirrhosis. However, due to its invasive nature, imaging modalities are commonly utilized for diagnosing different stages of NAFLD. Ultrasound and computed tomography (CT) are the most commonly used fist-line investigation with generally good sensitivity and specificity [10, 11, 12]. However, the sensitivity lowers when the level of steatosis is low [13]. Other techniques such as transient elastography MRI, MRI elastography, computer-assisted quantitative techniques have been developed to better assess steatosis and fibrosis [14, 15, 16, 17]. These techniques are not widely available and could associate with high cost. Simple biochemical markers such as low albumin, prolonged prothrombin time and thrombocytopenia should be incorporated into the diagnostic algorithm of NAFLD as well [1]. If a diagnosis is uncertain with imaging modalities or if there a high probability of liver fibrosis, a liver biopsy is warranted.

Advertisement

3. Epidemiology of NAFLD-associated HCC

3.1 Incidence of NAFLD-associated HCC

NAFLD is the fastest growing cause of HCC in the world. As only a small proportion of patients undergo screening and appropriate surveillance [18], it is extremely difficult to get representative estimates of HCC incidence in the general population with NAFLD. Thus, studies are limited to patients who underwent appropriate workup and surveillance. Given that diagnoses of the exact NAFLD stage can be challenging, the percentage of patients with NAFL, NASH and cirrhosis is unclear without definitive liver biopsy in most published studies. In one large population based study, the incidence of HCC in patients with NAFLD is estimated at 0.51% at year 12 [19]. The incidence of HCC increases as NAFLD progresses from NAFL to cirrhosis with the highest HCC incidence rate seen in those with NAFLD-associated liver cirrhosis. HCC incidence in patients with NAFL is low at 1.2 per 1000 person-years in a population-based study of US Veterans [20] and ranges from 0.3% to 0.43% in other studies that excluded patients with cirrhosis [19, 21, 22]. In contrast, HCC incidence in patients with cirrhosis dramatically rises to about 10 per 1000 person-years in the same US Veterans study [20], and could be up to 12.8% over 3 years in a systemic review [22].

NAFLD-associated HCC is becoming the leading cause of HCC as the prevalence of obesity and metabolic syndrome is rising. The increasing prevalence of NAFLD-associated HCC is reflected in studies utilizing large national transplant registries. Based on the data from the Scientific Registry of Transplant Recipients (SRTR) data system from 2002 to 2017 [23], 17% patients who were listed for liver transplant had a listing diagnosis of HCC. Even though chronic hepatitis C remained the leading cause of HCC in these transplant candidates and had a 6.2-fold increase from 2002 to 2017, NAFLD-associated HCC had an 11.5-fold increase. NAFLD-associated HCC is the most rapidly growing indication for liver transplantation. It comprised of 18% of liver transplant candidates with HCC in 2017 in contrast with that of 2% in 2002, while chronic hepatitis C related HCC decreased to 48% in 2017 from 53% in 2002. According to a predictive model, NAFLD prevalence among the adult population (aged ≥15 years) is projected at 33.5% in 2030; among the NAFLD cases, NASH cases are expected to increase from 20% to 27% by 2030; incidence of decompensated cirrhosis will increase 168% to 105,430 cases by 2030; prevalent HCC cases are expected to increase by 146% from 10,100 to 24,900 during 2015–2030; incident HCC cases are expected to increase by 137% from 5,160 to 12,240 in 2030 [9]. With more curative treatments being developed for chronic hepatitis C, the incidence of chronic hepatitis C related HCC is expected to decrease, while NAFLD-associated HCC is becoming the leading cause of HCC.

3.2 Risk factors of NAFLD-associated HCC

Many of the NAFLD risk factors including obesity, metabolic syndrome and T2DM are also independent risk factors of HCC. Other demographic risk factors including older age, male sex, Hispanic ethnicity, and genetic predisposition have also been studied as risk factors in HCC, including NAFLD-associated HCC.

3.2.1 Obesity, T2DM and metabolic syndrome

Obesity is closely related to both NAFLD and HCC. Obesity is the most common metabolic abnormality associated with NAFLD: an estimated 51.3% patients with NAFLD had obesity and up to 81.8% patients with NASH were obese [24]. Obesity is associated with many types of malignancies including HCC. It is assumed that 10% or more liver cancers could be attributable to excess weight [25]. T2DM is found in up to 71% of patients with NASH cirrhosis and is independently associated with increased risk of HCC [26]. A Surveillance, Epidemiology, and End Results (SEER)-Medicare based study from 1993 to 2005 showed that up to 37.1% patients with HCC had metabolic syndrome in contrast with 17.1% of comparison group residing in the same regions as the SEER registries. Among the metabolic conditions, T2DM or impaired fasting glucose has the strongest association with HCC. Metabolic syndrome remained significantly associated with increased risk of HCC even after adjusted multiple logistic regression analyses, suggesting metabolic syndrome as an independent risk factor for developing HCC [27].

3.2.2 Demographic and genetic risk factors

NAFLD-associated HCC present at a later age and is more prevalent in male. Significant racial and ethnic disparities in NAFLD and NAFLD-associated HCC in the United States are seen. Hispanics have the highest risk of developing NAFLD and NAFLD-associated HCC [28, 29]. Carriage of the PNPLA3 rs738409 C >G polymorphism is not only associated with greater risk of progressive steatohepatitis and fibrosis, but also of HCC with a 2.26-fold increased risk of HCC when carrying one copy of the allele, and 5-fold increased risk in homozygous individuals [30].

Advertisement

4. Pathogenesis of NAFLD associated HCC

NAFLD associated HCC is similar to many chronic liver disease related hepatocellular carcinomas, where hepatocellular injury and subsequent necroinflammation drive the formation of a protumorigenic microenvironment in the liver [31, 32, 33]. This microenvironment consists of a complex chronic inflammatory state with increased metabolic, oxidative, and mutagenic cellular stress, ultimately driving hepatocarcinogenesis (Figure 1) [31, 34, 35]. Different from other causes of HCC, e.g., viral or alcohol, an altered metabolomic state is the driver of hepatocellular injury in NAFLD associated HCC, playing an important role throughout the pathogenic process, and culminating in unique alterations of the molecular phenotype of the resulting tumor. The precise pathogenic mechanisms of NAFLD associated HCC continue to evolve, as the complex interplay of environmental factors, genetic susceptibility, and intricate inflammatory conditions further unfold.

Figure 1.

Stepwise pathogenesis of NAFLD associated HCC. In the setting of environmental and genetic predisposition, the sequalae of metabolic reprograming and hepatocellular injury in NAFLD lead to the creation of a protumorigenic microenvironment and ultimately hepatocarcinogenesis. Created with BioRender.com.

4.1 Hepatocellular injury and the production of a protumorigenic microenvironment

The original model of hepatocellular injury in NASH was described as a “two-hit” hypothesis, where the first hit, steatosis, sensitizes the hepatocyte to injury and cell death resulting from the “second-hit” of oxidative stress [36]. Although this model is largely seen as overly reductive, it does provide a conceptual framework for hepatocellular injury in NAFLD associated HCC. Lipotoxicity, a state of lipid dysregulation leading to organelle dysfunction and cell death, is often-considered the initial metabolic insult that causes hepatocellular injury in NAFLD [32]. Increased hepatic lipid deposits are a hallmark of NAFLD. Multiple features of metabolic syndrome, including excess dietary free fatty acids (FFAs), excess FFA release from adipose tissue, increased insulin resistance, upregulated de-novo lipogenesis and alterations of the gut microbiota all contribute to a lipid-rich hepatic metabolic state [37]. At increased concentrations in the liver, lipids are directly and indirectly hepatotoxic, promoting proapoptotic and ER-stress pathways, while inducing mitochondrial dysfunction and reactive oxygen species (ROS) production [38]. In addition to lipid induced hepatocellular injury, derangements in other metabolic pathways seen in NAFLD, including bile acid metabolism and iron storage, likely contribute to hepatocyte damage and hepatocarcinogenesis as well [39, 40].

As in many cancers, the innate and adaptive immune system play a Janus-faced role in tumor development: both promoting necroinflammation and therefore carcinogenesis, while also performing antitumor cell killing and immune surveillance [41]. Hepatocellular injury in NAFLD plays a central role in disrupting this balancing act, skewing the immune response to favor tumorigenesis. Hepatocyte death stimulates the immune response through exposure of immune cells to damage associated molecular patterns (DAMPS). Additionally, microbiome changes in NAFLD patients likely contribute to the inflammatory immune phenotype, with the increased “leakiness” gut leading to increased translocation of lipopolysaccharide (LPS) and other pathogen associated molecular patterns (PAMPS) into the portal circulation [42]. Both DAMPS and PAMPS act through Toll-like receptors (TLR) and other pattern recognition receptors to activate liver resident macrophages (Kupffer cells). Activated Kupffer cells (KCs) recruit and stimulate other innate and adaptive immune cell subsets which secrete proinflammatory factors including IL-1β, IL-2, IL-7, IL-12, IL-15, TNFα, and IFNγ, further promoting an immunostimulatory and cytotoxic environment. A major consequence of the immunostimulatory environment is the activation of non-parenchymal hepatic cells including hepatic stellate cells, which increase extracellular matrix deposition and fibrosis. Moreover, stimulated innate cells directly contribute to the abundance of ROS and therefore oxidative DNA damage in the liver due to increased respiratory burst activity [41]. Together these immunostimulatory processes perpetuate hepatic cell death, promote hepatic stellate cell mediated fibrosis, and contribute genotoxic metabolites to the microenvironment, all key drivers of hepatocarcinogenesis. In response to these chronic inflammatory conditions, many immune exhaustion responses are induced, including the expression of immunosuppressive factors (IL-10, TGFβ) and the immune checkpoint PD-L1. While this immunosuppressive response contributes to the reduction of detrimental inflammation, antitumor cytotoxic immune response is inhibited as well, contributing to tumor growth.

In addition to the directly cytotoxic and genotoxic mechanisms described above, the positive feedback loop of hepatocellular injury, necroinflammation and fibrosis indirectly promote tumor progression through induction of angiogenesis. In response to inflammatory stimuli, activated monocytes increase production of VEGF and MMP9, promoting tumor neovascularization, growth, and metastasis [43]. Notably even prior to HCC development, NAFLD patients exhibit increased serologic markers of angiogenesis and increased neovascularization in biopsy samples [44], further contributing to the confluence of protumorigenic factors ultimately leading to tumorigenesis in NAFLD.

The influence of metabolic syndrome can be observed in each of these protumorigenic mechanisms, hepatocellular injury, chronic inflammation, immune exhaustion, and increased neovascularization. Many features of metabolic syndrome including hyperlipidemia, hyperglyceridemia, and obesity directly contribute to steatosis and lipotoxic hepatocellular injury. Adipocytes directly produce multiple inflammatory cytokines (TNFα, IL-6) and proangiogenic factors (VEGF, FDGF), likely contributing to oncogenic chronic inflammation, immune exhaustion, and angiogenesis [45]. Mouse models of NAFLD induced HCC highlight the importance of metabolic syndrome in HCC pathogenesis. In mice with diet ± activity modifications designed to recapitulate conditions common metabolic syndrome, the vast majority (60–89%) of mice develop HCC [46], suggesting a potent role of metabolic syndrome in HCC development.

4.2 Hepatocarcinogenesis and disease progression

Ultimately the protumorigenic microenvironment results in DNA damage and subsequent mutagenesis. DNA oxidative damage is a major contributor to mutagenesis in NAFLD associated HCC. The DNA oxidative stress marker 8-hydroxy-2′-deoxyguanosine (8-OHdG) in NAFLD associated HCC is increased compared to that of healthy patient livers or tumors from patients with viral and alcohol associated HCC [47]. 8-OHdG is an independent risk factor for hepatocarcinogenesis and therefore highlights the role of oxidative damage in HCC pathogenesis. In addition to genotoxic alterations from DNA oxidative damage, oxidative damage can cause epigenetic changes, which may play a role in HCC carcinogenesis. Epigenetic inactivation of tumor suppressor genes consistent with oxidative DNA damage response have been observed in NAFLD induced HCC patients [48]. Furthermore, alterations in DNA repair pathways may also contribute to genomic instability. Upregulation of DNA-dependent protein kinase, a central member of the error prone DNA repair mechanism non-homologous end joining (NHEJ), has been observed in NAFLD associated HCC [49]. Together these mechanisms lead to an increased mutagenic state in NAFLD associated HCC.

Although a wide variety of mutations have been documented in NAFLD associated HCC, hotspot genes and mutational signatures have been described. In a cohort of 80 patients with NAFLD associated HCC, the most frequently mutated genes were the telomerase (TERT) promoter (56%); the gene encoding beta-catenin, CTNNB1 (28%); the tumor suppressor, TP53 (18%); and the activin receptor, ACVR2A (10%) [35]. Notably, TERT promotor, CTNNB1 and TP53 are mutated at similar rates in HCC patients en masse regardless of etiology; however, mutations in ACVR2A are more enriched in patients with NAFLD associated HCC compared with that in other etiologies [50]. Transcriptionally, the majority of NAFLD-associated HCC tumors demonstrated upregulation of either the WNT–TGFβ or WNT–β-catenin oncogenic signaling pathways, highlighting the importance of both non-canonical and canonical WNT signaling in NAFLD associated HCC carcinogenesis [35]. Moreover, other transcriptional signatures consistent with underlying pathogenic features of NAFLD associated HCC are enriched in these patients including bile acid metabolism, oxidative stress, and inflammation-related gene signatures. Together these genomic and transcriptomic alterations drive malignant transformation and disease progression.

Advertisement

5. Current treatment of NAFLD-associated HCC

Current HCC treatment recommendations incorporate Barcelona Clinic Liver Cancer (BCLC) stage, liver lesion number and size, liver function and patient performance status [51]. Treatment modalities for localized disease include hepatic resection, ablation, and liver transplant for single or small lesions; chemoembolization and stereotactic radiation for larger, multiple unresectable lesions. These locoregional treatments seem to be equally effective regardless of HCC etiology. A comprehensive review on different locoregional treatment modalities in NAFLD-associated HCC has been published recently [52]. This chapter will focus on systemic therapies, especially immune checkpoint inhibitors in NAFLD-associated HCC.

5.1 Tyrosine kinase inhibitor (TKI)

5.1.1 Sorafenib

The Phase III SHARP trial established the efficacy of sorafenib, a multikinase inhibitor, which prolonged the median survival and median time to radiographic progression for patients with locally advanced or metastatic HCC [53]. A similar Phase III trial that was conducted in Asian-Pacific region observed similar results [54]. In the SHARP study, 48% patients had viral hepatitis as an etiology of HCC and the percentage of NAFLD-associated HCC is unknown. The subsequent published subgroup analysis of SHARP study patients and combined analysis of SHARP study and Asian-Pacific study patients didn’t include NAFLD-associated HCC as a subgroup either [55, 56]. A recent presented study of 5201 patients with HCC treated with sorafenib found that there is no overall survival or adverse event difference between NAFLD-associated HCC and other etiology related HCC. However, patients with NAFLD-associated HCC were significantly older and sorafenib was commenced at more advanced stages. There were only 3.6% patients with NAFLD-associated HCC in the study, limiting the ability of drawing definite conclusions [57].

5.1.2 Lenvatinib

Lenvatinib, another multikinase inhibitor, showed non-inferiority to sorafenib in the Phase III REFLECT study [58]. Similar to SHARP study, patients in REFLECT study was characterized as having viral etiologic HCC or alcohol related HCC. The percentage of NALFD-associated HCC is unknown. A recently published multi-center retrospective study from Japan included 530 HCC patients treated with Lenvatinib [59]. The study compared the survival of 103 patients with NAFLD-associated HCC with that of 427 patients with HCC from other etiologies and revealed that progression free survival was statistically better in patients with NAFLD-associated HCC (9.3 vs. 7.5 months, P = 0.012), and overall survival was numerically better even though not statistically significant (20.5 vs. 16.9 months, P = 0.057).

5.1.3 Other TKIs

Regorafenib was approved as a second-line treatment for advanced HCC after progressing on sorafenib based on the Phase III RESORCE study [60]. Ramucirumab was also approved for the same indication after showing overall survival and progression free survival benefit in patients with advanced HCC in REACH-2 study [61]. Multikinase inhibitor cabozantinib improved survival and PFS in patients who have failed one or two lines of treatment in the study CELESTIAL and cabozantinib was the first approved third-line treatment for advanced HCC [62]. In these three studies, subgroup analysis didn’t show different response to these TKIs according to HCC etiology. In a retrospective study that included 23.5% NAFLD-associated HCC also showed similar responses to regorafenib regardless of HCC etiologies. However, there were less than 10% of NAFLD-associated HCC patients included in RESORCE, REACH-2 and CELESTIAL studies. And there were less than 25 patients in the retrospective study mentioned above. The small number of patients enrolled in the studies limit definitive conclusions.

5.2 Immune checkpoint inhibitors

Checkpoint inhibitor single agent was first studied in the second line settings. Both nivolumab and pembrolizumab showed improved response rate in the early phase clinical trials [63, 64]. Based on the results from these studies, FDA granted accelerated approval for advanced HCC. Pembrolizumab was further tested in the second line setting in KEYNOTE 240 study. However, the study didn’t meet the primary endpoint of superior combined OS and PFS [65]. Similarly, Phase III CheckMate 459 study comparing nivolumab with sorafenib in the first line setting didn’t show superiority of nivolumab [66]. Combing nivolumab and ipilimumab at different dose and schedule was explored and doublet regimen has shown improved response rate up to 30% in the second line setting [67]. Phase III HIMALAYA study added tremelimumab to durvalumab and compared this regimen with sorafenib. The combination treatment improved overall survival compared to sorafenib and the final results are eagerly anticipated [68].

As with studies of TKIs in HCC, most studies of immune checkpoint inhibitors stratified patients to groups of HBV related, HCV related and uninfected, and the percentage of NAFLD-associated HCC in the uninfected group is unknown. Nevertheless, subgroup analysis in most of these studies showed comparable efficacy of immune checkpoint inhibitors in uninfected and overall population (Table 1).

Study name/phase of studyDrug(s)Etiology and patient numbersEndpoint and results
Single agent
CheckMate 040/Phase I/IINivolumabN = 262
HBV = 25.2%
HCV = 22.9%
Uninfected = 51.9%
ORR (CR+PR) 20%
HBV 14%
HCV 20%
Uninfected 21–23%
CheckMate 459/Phase IIINivolumab vs.
sorafenib
N = 743 (371 vs. 372)
HBV = 31%
HCV = 23%
Uninfected = 45%
OS 16.4m vs. 14.7m
ORR (CR+PR) 15% vs. 7%
HBV 19% vs. 8%
HCV 17% vs. 7%
Uninfected 12% vs. 7%
Keynote 224/Phase IIPembrolizumabN = 104
HBV 21%
HCV 25%
Uninfected 33%
ORR (CR+PR) 18.3%
Infected (HBV+HCV) 13%
Uninfected 20%
Keynote 240/Phase IIIPembrolizumab vs. BSCN = 413
HBV 21.5–25.9%
HCV 15.5–15.6%
Uninfected 58.6–63.0%
OS and PFS*
OS 13.9m vs. 10.6 m
PFS 3.0m vs. 2.8 m
Doublet
HIMALAYA/Phase IIITremelimumab+durvalumab vs. sorafenibN = 1171
HBV 30.6–31%
HCV 27.5–28%
Nonviral 41–41.9%
OS 16.43 m vs. 13.77 m
CheckMate 040/Phase I/IINivolumab + ipilimumab#N = 148
HBV 51%
HCV 22%
Uninfected 22%
ORR (CR+PR) ^
27–32%

Table 1.

Overview of outcomes of checkpoint inhibitor studies in patients with locally advanced or metastatic hepatocellular carcinoma.

OS hazard ratios in patients with HBV, HCV and uninfected were 0.57 (0.35–0.94), 0.96 (0.48–1.92) and 0.88 (0.64–1.20) respectively. PFS hazard ratios in patients with HBV, HCV and uninfected were 0.70 (0.44–1.13), 0.46 (0.24–0.90), 0.75 (0.56–1.01).


Arm A: nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, administered every 3 weeks (4 doses), followed by nivolumab 240 mg every 2 weeks; Arm B: nivolumab 3 mg/kg plus ipilimumab 1 mg/kg, administered every 3 weeks (4 doses), followed by nivolumab 240 mg every 2 weeks; Arm C: nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks (arm C).


Median overall survival of patients who were HBV/HCV uninfected, HBV infected, or HCV infected in arm A was 22.2 months, 22.8 months, and 14.9 months; in arm B, 11.8 months, 12.1 months, and 16.1 months; and in arm C, 7.4 months, 9.6 months, and 33.0 months, respectively.


Abbreviations: HBV: hepatitis B; HCV: hepatitis C; BSC: best supportive care; ORR: objective response rate; CR: complete remission; PR: partial remission; OS: overall survival; PFS: progression free survival.

5.3 Immune checkpoint inhibitor and VGEF inhibitor

The combination of immune checkpoint inhibitor atezolizumab and VEGF inhibitor bevacizumab in patients with locally advanced or metastatic HCC in the first setting has shown superior OS and PFS compared to sorafenib in IMbrave150 trial [69]. The median OS was 19.2 months with atezolizumab plus bevacizumab and 13.4 months with sorafenib (hazard ratio 0.66; 95% CI 0.52–0.85; p <0.001). The median PFS was 6.9 and 4.3 months in the respective treatment groups (HR 0.65; 95% CI 0.53–0.81; p < 0.001). The combination was approved by FDA as first line treatment for HCC in 2020 and is the preferred first line treatment over single agent immune checkpoint inhibitor or TKIs in eligible patients.

The percentage of NAFLD-associated HCC is unknown in this study but 30% of the patients had no viral infection. In the subgroup analysis, uninfected patients with HCC seemed to have derived less benefit from the combination treatment compared to patients with viral hepatitis (HR for death 1.05; 95% CI 0.68–1.63) [70]. A previous animal study has demonstrated the existence of a CD8+PD-1+ subset of protumorigenic cells in NASH that favor the development of HCC and hamper response to immune checkpoint inhibitors. The authors also performed meta-analysis using data from CheckMate 459, IMbrave150 and KEYNOTE240, and revealed that patients with non-viral etiology had inferior survival when treated with immune checkpoint inhibitors. They then performed a cohort study of 130 patients with HCC from various etiologies and patients with NAFLD associated HCC had shorter overall survival of 5.4 months vs. 11 months in patients with HCC from other etiologies.

The better response to immunotherapy in patients with viral-induced HCC than in patients with NAFLD associated HCC might be due to the amount or quality of viral antigens or to a different liver micro-environment, possibly one that does not impair immune surveillance. Stratifying clinical trials according to HCC etiology for prospective validation in future clinical trials is warranted.

Advertisement

6. Conclusions

As incidence of HBV and HCV associated HCC continues to decrease with effective antiviral treatment, NAFLD associated HCC is becoming the leading cause of HCC. In light of the rising prevalence of NAFLD associated HCC, its unique pathogenesis and findings suggestive inferior response to immune checkpoint inhibitors, future clinical trials should assess whether HCC etiology influence the efficacy of a given treatment.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Jennison E, Patel J, Scorletti E, Byrne CD. Diagnosis and management of non-alcoholic fatty liver disease. Postgraduate Medical Journal. 2019;95(1124):314-322
  2. 2. Wong VW, Wong GL, Choi PC, Chan AW, Li MK, Chan HY, et al. Disease progression of non-alcoholic fatty liver disease: A prospective study with paired liver biopsies at 3 years. Gut. 2010;59(7):969-974
  3. 3. McPherson S, Hardy T, Henderson E, Burt AD, Day CP, Anstee QM. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: Implications for prognosis and clinical management. Journal of Hepatology. 2015;62(5):1148-1155
  4. 4. Ekstedt M, Franzen LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology. 2006;44(4):865-873
  5. 5. Calzadilla Bertot L, Adams LA. The natural course of non-alcoholic fatty liver disease. International Journal of Molecular Science. 2016;17:5
  6. 6. Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, Loomba R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: A systematic review and meta-analysis of paired-biopsy studies. Clinical Gastroenterology and Hepatology. 2015;13:643
  7. 7. Pais R, Charlotte F, Fedchuk L, Bedossa P, Lebray P, Poynard T, et al. A systematic review of follow-up biopsies reveals disease progression in patients with non-alcoholic fatty liver. Journal of Hepatology. 2013;59(3):550-556
  8. 8. Ward ZJ, Bleich SN, Cradock AL, Barrett JL, Giles CM, Flax C, et al. Projected U.S. State-level prevalence of adult obesity and severe obesity. The New England Journal of Medicine. 2019;381(25):2440-2450
  9. 9. Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology. 2018;67(1):123-133
  10. 10. Lee SS, Park SH, Kim HJ, Kim SY, Kim M-Y, Kim DY, et al. Non-invasive assessment of hepatic steatosis: Prospective comparison of the accuracy of imaging examinations. Journal of Hepatology. 2010;52(4):579-585
  11. 11. Hernaez R, Lazo M, Bonekamp S, Kamel I, Brancati FL, Guallar E, et al. Diagnostic accuracy and reliability of ultrasonography for the detection of fatty liver: A meta-analysis. Hepatology. 2011;54(3):1082-1090
  12. 12. Ballestri S, Nascimbeni F, Baldelli E, Marrazzo A, Romagnoli D, Targher G, et al. Ultrasonographic fatty liver indicator detects mild steatosis and correlates with metabolic/histological parameters in various liver diseases. Metabolism. 2017;72:57-65
  13. 13. Saadeh S, Younossi ZM, Remer EM, Gramlich T, Ong JP, Hurley M, et al. The utility of radiological imaging in nonalcoholic fatty liver disease. Gastroenterology. 2002;123(3):745-750
  14. 14. Talwalkar JA, Kurtz DM, Schoenleber SJ, West CP, Montori VM. Ultrasound-based transient elastography for the detection of hepatic fibrosis: Systematic review and meta-analysis. Clinical Gastroenterology and Hepatology. 2007;5(10):1214-1220
  15. 15. Noureddin M, Lam J, Peterson MR, Middleton M, Hamilton G, Le T-A, et al. Utility of magnetic resonance imaging versus histology for quantifying changes in liver fat in nonalcoholic fatty liver disease trials. Hepatology. 2013;58(6):1930-1940
  16. 16. Zhang B, Ding F, Chen T, Xia L-H, Qian J, Lv G-Y. Ultrasound hepatic/renal ratio and hepatic attenuation rate for quantifying liver fat content. World Journal of Gastroenterology. 2014;20(47):17985-17992
  17. 17. Loomba R, Wolfson T, Ang B, Hooker J, Behling C, Peterson M, et al. Magnetic resonance elastography predicts advanced fibrosis in patients with nonalcoholic fatty liver disease: A prospective study. Hepatology. 2014;60(6):1920-1928
  18. 18. Ioannou GN. Epidemiology and risk-stratification of NAFLD-associated HCC. Journal of Hepatology. 2021;75(6):1476-1484
  19. 19. Kawamura Y, Arase Y, Ikeda K, Seko Y, Imai N, Hosaka T, et al. Large-Scale Long-Term Follow-Up Study of Japanese patients with non-alcoholic fatty liver disease for the onset of hepatocellular carcinoma. The American Journal of Gastroenterology. 2012;107(2):253-261
  20. 20. Kanwal F, Kramer JR, Mapakshi S, Natarajan Y, Chayanupatkul M, Richardson PA, et al. Risk of hepatocellular cancer in patients with non-alcoholic fatty liver disease. Gastroenterology. 2018;155(6):1828
  21. 21. Sørensen HT, Mellemkjaer L, Jepsen P, Thulstrup AM, Baron J, Olsen JH, et al. Risk of cancer in patients hospitalized with fatty liver: A Danish cohort study. Journal of Clinical Gastroenterology. 2003;36(4):356-359
  22. 22. White DL, Kanwal F. Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clinical Gastroenterology and Hepatology. 2012;10(12):1342
  23. 23. Younossi Z, Stepanova M, Ong JP, Jacobson IM, Bugianesi E, Duseja A, et al. Nonalcoholic steatohepatitis is the fastest growing cause of hepatocellular carcinoma in liver transplant candidates. Clinical Gastroenterology and Hepatology. 2019;17(4):748
  24. 24. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73-84
  25. 25. Bhaskaran K, Dos-Santos-Silva I, Leon DA, Douglas IJ, Smeeth L. Association of BMI with overall and cause-specific mortality: A population-based cohort study of 3·6 million adults in the UK. The Lancet Diabetes and Endocrinology. 2018;6(12):944-953
  26. 26. Yang JD, Ahmed F, Mara KC, Addissie BD, Allen AM, Gores GJ, et al. Diabetes is associated with increased risk of hepatocellular carcinoma in patients with cirrhosis from nonalcoholic fatty liver disease. Hepatology. 2020;71(3):907-916
  27. 27. Welzel TM, Graubard BI, Zeuzem S, El-Serag HB, Davila JA, McGlynn KA. Metabolic syndrome increases the risk of primary liver cancer in the United States: A study in the SEER-Medicare database. Hepatology. 2011;54(2):463-471
  28. 28. Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contreras M, et al. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: A prospective study. Gastroenterology. 2011;140(1):124-131
  29. 29. Rich NE, Hester C, Odewole M, Murphy CC, Parikh ND, Marrero JA, et al. Racial and ethnic differences in presentation and outcomes of hepatocellular carcinoma. Clinical Gastroenterology and Hepatology. 2019;17(3):551
  30. 30. Liu YL, Patman GL, Leathart JB, Piguet AC, Burt AD, Dufour JF, et al. Carriage of the PNPLA3 rs738409 C >G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma. Journal of Hepatology. 2014;61(1):75-81
  31. 31. Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: Current concepts and future challenges. Nature Reviews. Gastroenterology & Hepatology. 2019;16(7):411-428
  32. 32. Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. Journal of Hepatology. 2018;68(2):280-295
  33. 33. Hatting M, Tacke F. From NAFLD to HCC: Is IL-17 the crucial link? Hepatology. 2017;65(2):739-741
  34. 34. Huang DQ , El-Serag HB, Loomba R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nature Reviews. Gastroenterology & Hepatology. 2021;18(4):223-238
  35. 35. Pinyol R, Torrecilla S, Wang H, Montironi C, Pique-Gili M, Torres-Martin M, et al. Molecular characterisation of hepatocellular carcinoma in patients with non-alcoholic steatohepatitis. Journal of Hepatology. 2021;75(4):865-878
  36. 36. Day CP, James OF. Steatohepatitis: A tale of two “hits”? Gastroenterology. 1998;114(4):842-845
  37. 37. Lambert JE, Ramos-Roman MA, Browning JD, Parks EJ. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014;146(3):726-735
  38. 38. Sunny NE, Bril F, Cusi K. Mitochondrial adaptation in nonalcoholic fatty liver disease: Novel mechanisms and treatment strategies. Trends in Endocrinology and Metabolism. 2017;28(4):250-260
  39. 39. Sydor S, Best J, Messerschmidt I, Manka P, Vilchez-Vargas R, Brodesser S, et al. Altered microbiota diversity and bile acid signaling in cirrhotic and noncirrhotic NASH-HCC. Clinical and Translational Gastroenterology. 2020;11(3):e00131
  40. 40. Yu YC, Luu HN, Wang R, Thomas CE, Glynn NW, Youk AO, et al. Serum biomarkers of iron status and risk of hepatocellular carcinoma development in patients with nonalcoholic fatty liver disease. Cancer Epidemiology, Biomarkers & Prevention. 2022;31(1):230-235
  41. 41. Ringelhan M, Pfister D, O’Connor T, Pikarsky E, Heikenwalder M. The immunology of hepatocellular carcinoma. Nature Immunology. 2018;19(3):222-232
  42. 42. Luther J, Garber JJ, Khalili H, Dave M, Bale SS, Jindal R, et al. Hepatic injury in nonalcoholic steatohepatitis contributes to altered intestinal permeability. Cellular and Molecular Gastroenterology and Hepatology. 2015;1(2):222-232
  43. 43. Ehling J, Bartneck M, Wei X, Gremse F, Fech V, Mockel D, et al. CCL2-dependent infiltrating macrophages promote angiogenesis in progressive liver fibrosis. Gut. 2014;63(12):1960-1971
  44. 44. Lei L, Ei Mourabit H, Housset C, Cadoret A, Lemoinne S. Role of angiogenesis in the pathogenesis of NAFLD. Journal of Clinical Medicine. 2021;10(7):1960-1971
  45. 45. Cabia B, Andrade S, Carreira MC, Casanueva FF, Crujeiras AB. A role for novel adipose tissue-secreted factors in obesity-related carcinogenesis. Obesity Reviews. 2016;17(4):361-376
  46. 46. Carlessi R, Kohn-Gaone J, Olynyk JK, Tirnitz-Parker JEE. Mouse models of hepatocellular carcinoma. In: Tirnitz-Parker JEE, editor. Hepatocellular Carcinoma. Brisbane (AU): Codon Publications; 2019
  47. 47. Tanaka S, Miyanishi K, Kobune M, Kawano Y, Hoki T, Kubo T, et al. Increased hepatic oxidative DNA damage in patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. Journal of Gastroenterology. 2013;48(11):1249-1258
  48. 48. Nishida N, Yada N, Hagiwara S, Sakurai T, Kitano M, Kudo M. Unique features associated with hepatic oxidative DNA damage and DNA methylation in non-alcoholic fatty liver disease. Journal of Gastroenterology and Hepatology. 2016;31(9):1646-1653
  49. 49. Cornell L, Munck JM, Alsinet C, Villanueva A, Ogle L, Willoughby CE, et al. DNA-PK-A candidate driver of hepatocarcinogenesis and tissue biomarker that predicts response to treatment and survival. Clinical Cancer Research. 2015;21(4):925-933
  50. 50. Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology. 2015;149(5):1226
  51. 51. European Association for the Study of the Liver. Electronic address eee, European Association for the Study of the L. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. Journal of Hepatology. 2018;69(1):182-236
  52. 52. Foerster F, Gairing SJ, Müller L, Galle PR. NAFLD-driven HCC: Safety and efficacy of current and emerging treatment options. Journal of Hepatology. 2022;76(2):446-457
  53. 53. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J-F, et al. Sorafenib in advanced hepatocellular carcinoma. The New England Journal of Medicine. 2008;359(4):378-390
  54. 54. Cheng A-L, Kang Y-K, Chen Z, Tsao C-J, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. The Lancet Oncology. 2009;10(1):25-34
  55. 55. Bruix J, Raoul J-L, Sherman M, Mazzaferro V, Bolondi L, Craxi A, et al. Efficacy and safety of sorafenib in patients with advanced hepatocellular carcinoma: Subanalyses of a phase III trial. Journal of Hepatology. 2012;57(4):821-829
  56. 56. Bruix J, Cheng A-L, Meinhardt G, Nakajima K, De Sanctis Y, Llovet J. Prognostic factors and predictors of sorafenib benefit in patients with hepatocellular carcinoma: Analysis of two phase III studies. Journal of Hepatology. 2017;67(5):999-1008
  57. 57. Howell J, Samani A, Mannan B, Hajiev S, Aval LM, Abdelmalak R, et al. Impact of NAFLD on clinical outcomes in hepatocellular carcinoma treated with sorafenib: An international cohort study. JCO. 2021;39(3):289
  58. 58. Kudo M, Finn RS, Qin S, Han K-H, 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. The Lancet. 2018;391(10126):1163-1173
  59. 59. Hiraoka A, Kumada T, Tada T, Tani J, Kariyama K, Fukunishi S, et al. Efficacy of lenvatinib for unresectable hepatocellular carcinoma based on background liver disease etiology: Multi-center retrospective study. Scientific Reports. 2021;11(1):16663
  60. 60. Bruix J, Qin S, Merle P, Granito A, Huang Y-H, 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. The Lancet. 2017;389(10064):56-66
  61. 61. Zhu AX, Park JO, Ryoo B-Y, Yen C-J, Poon R, Pastorelli D, et al. Ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma following first-line therapy with sorafenib (REACH): A randomised, double-blind, multicentre, phase 3 trial. The Lancet Oncology. 2015;16(7):859-870
  62. 62. Abou-Alfa GK, Meyer T, Cheng A-L, El-Khoueiry AB, Rimassa L, Ryoo B-Y, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. The New England Journal of Medicine. 2018;379(1):54-63
  63. 63. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet. 2017;389(10088):2492-2502
  64. 64. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. The Lancet Oncology. 2018;19(7):940-952
  65. 65. Finn RS, Ryoo B-Y, Merle P, Kudo M, Bouattour M, Lim HY, et al. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. Journal of Clinical Oncology. 2020;38(3):193-202
  66. 66. Yau T, Park J-W, Finn RS, Cheng A-L, Mathurin P, Edeline J, et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. The Lancet Oncology. 2022;23(1):77-90
  67. 67. Yau T, Kang Y-K, Kim T-Y, El-Khoueiry AB, Santoro A, Sangro B, et al. Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncology. 2020;6(11):e204564
  68. 68. Dual Immunotherapy Makes Strides against HCC. Cancer Discovery. 2022;12(4). Available from: https://aacrjournals.org/cancerdiscovery/article/12/4/OF1/689618/Dual-Immunotherapy-Makes-Strides-against-HCCDual
  69. 69. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim T-Y, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. The New England Journal of Medicine. 2020;382(20):1894-1905
  70. 70. Pfister D, Nunez NG, Pinyol R, Govaere O, Pinter M, Szydlowska M, et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature. 2021;592(7854):450-456

Written By

Kai Sun, Alan Hodges and Maen Abdelrahim

Submitted: 26 June 2022 Reviewed: 27 July 2022 Published: 13 April 2023