Next Article in Journal
Pancreatic Cancer Education: A Scoping Review of Evidence Across Patients, Professionals and the Public
Previous Article in Journal
Machine Learning in Biomarker-Driven Precision Oncology: Automated Immunohistochemistry Scoring and Emerging Directions in Genitourinary Cancers
Previous Article in Special Issue
STAT2 Promotes Tumor Growth in Colorectal Cancer Independent of Type I IFN Receptor Signaling
error_outline You can access the new MDPI.com website here. Explore and share your feedback with us.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hepatocellular Carcinoma Around the Clock

by
Mariana Verdelho Machado
1,2,3
1
Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
2
Unidade de Oncologia Digestiva, Fundação Champalimaud, 1400-038 Lisbon, Portugal
3
Serviço de Gastrenterologia, Hospital de Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
Curr. Oncol. 2026, 33(1), 32; https://doi.org/10.3390/curroncol33010032
Submission received: 30 November 2025 / Revised: 28 December 2025 / Accepted: 5 January 2026 / Published: 7 January 2026

Simple Summary

Lifestyle-related circadian rhythm disruptions favor the development of hepatocellular carcinoma (HCC) both indirectly (by promoting precursor conditions such as metabolic dysfunction-associated liver disease and liver cirrhosis) and directly through intrinsic oncogenic effects. Population-level interventions that promote circadian realignment may help prevent the increasing incidence of HCC. Assessing circadian disruption could also help determine the prognosis of individual patients and their potential responses to therapy. Therapy strategies should incorporate chronotherapy principles, aiming not only to realign patient’s circadian cycles but also to optimize the timing of therapeutic delivery according to circadian variations in pharmacokinetics and pharmacodynamics to improve efficacy.

Abstract

The dramatic shift in human behavior from hunter-gatherer to modern lifestyles has led to a systematic disruption of the human circadian cycle. Contributors include night-shift work, jet lag, and less intuitive but widespread factors, such as exposure to artificial light at night and irregular eating schedules. Circadian disruption is classified as a Group 2A carcinogen by the International Agency for Research on Cancer (IARC). Hepatocellular carcinoma (HCC) is the third most deadly cancer worldwide, with a rising prevalence in Western countries, largely driven by increasing rates of obesity and steatotic liver disease-associated hepatocarcinogenesis. Emerging evidence suggests that circadian disruption plays a significant role in HCC pathogenesis. Several genes involved in metabolism, cell survival, and immunosurveillance are under the control of circadian rhythms. Experimental preclinical data and epidemiological studies have indicated a strong association between circadian disruption and HCC development. Moreover, molecular signatures related to circadian regulation appear to accurately predict the prognosis of patients with HCC. The concept of chronotherapy is also gaining interest, with studies suggesting improved immunotherapy effectiveness when immune checkpoint inhibitors are administered in the morning. This review summarizes the current literature on the impact of circadian disruption on HCC pathogenesis, prognosis, and treatment.

1. Introduction

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide [1]. This mortality ranking contrasts with its position as the sixth most common cancer globally, reflecting the disease’s poor survival rates, with fewer than 20% of patients alive five years after diagnosis [2]. These statistics are even more concerning given predictions that, over the next 20 years, both the incidence and mortality of HCC will increase by approximately 50% [3]. The high mortality associated with HCC is due to both late diagnosis (with only 15–20% of patients being diagnosed at early, potentially curable stages) and the tumor’s aggressiveness and low treatment response rates [4,5]. The epidemiology of HCC etiology is also shifting: while chronic viral hepatitis is declining, steatotic liver disease (SLD), metabolic dysfunction-associated (MASLD) and alcohol-associated, has become the fastest growing cause of HCC [6]. As a result, lifestyle-related factors now account for approximately 52% of the attributable risk for HCC, including alcohol use, tobacco smoking, poor diet quality, and physical inactivity [7]. Lifestyle factors that disrupt circadian rhythms, such as abnormal sleep duration and night-shift work, may also significantly impact HCC epidemiology [8].
Indeed, “night-shift work”, and “shift work involving circadian disruption”, have been considered probable carcinogenics (Group 2A) by the International Agency for Research on Cancer (IARC) in 2007 and reaffirmed in 2019 [9].
The word circadian comes from the Latin circa (approximately) and diem (day). Most organisms, including animals and plants, exhibit circadian rhythms, reflecting their need to adapt to the environmental changes produced by the daily light–dark cycle generated by the Earth’s rotation around its axis [10]. Virtually all organs and cells display circadian rhythms that are hierarchically regulated by a central clock in the suprachiasmatic nuclei. Importantly, most physiological functions are under circadian control, including molecular mechanisms that, when disturbed, may contribute to cancer development, such as cell proliferation and differentiation, DNA damage repair, apoptosis, autophagy, anabolic and catabolic processes, and oxidative stress [11]. In the liver, approximately 15% of the transcriptome is regulated by circadian genes [12].
There is a significant amount of evidence from preclinical, epidemiological, and clinical research suggesting a strong association between circadian rhythm disruption and HCC development and poor prognosis. Furthermore, growing evidence suggests that chronotherapy may affect the treatment response to radiation and immunotherapy in patients with HCC.
This review summarizes the clinical evidence on the impact of circadian rhythms on the development, treatment, and prognosis of HCC and describes the potential mechanisms underlying these associations.

2. Physiology of the Circadian Rhythm

The maestro regulating the circadian rhythm is the central clock, located in the suprachiasmatic nuclei, which responds to external cues or zeitgebers, such as light, food intake, physical activity, and temperature [13]. Although peripheral clocks are under the control of the central clock via neuronal signals (mainly the autonomic nervous system), hormonal signals (e.g., cortisol and melatonin), and metabolic cues, they can also autonomously entrain circadian rhythms [14].
Rhythmicity is maintained by the oscillatory expression of several transcription factors, achieved through autoregulatory transcription loops [15]. At the top of this molecular hierarchy are two transcription factors that heterodimerize: circadian locomotor output cycle kaput (CLOCK) and brain and muscle ARNT-like-1 (BMAL1) [16]. At the start of the circadian day, the CLOCK-BMAL1 complex activates transcription by binding to E-box enhancer elements in the promoters of thousands of target genes [17], including period family genes (PER1, PER2, and PER3) [18], and cryptochrome family genes (CRY1 and CRY2) [19].
PER and CRY proteins heterodimerize and act as transcription repressors, inhibiting CLOCK-BMAL1 activity at night [20]. They are subsequently phosphorylated, PER proteins by casein kinase 1 (CK1) [21] and CRY proteins by other kinases such as AMPK [22], and ubiquitinated by E3 ligases, targeting them for proteasomal degradation [23]. This process restores CLOCK-BMAL1 activity, initiating a new cycle [20].
Other CLOCK-BMAL1 targets include retinoic acid-related orphan nuclear receptors (ROR) and REV-ERB. RORs act as transcriptional activators, whereas REV-ERBs act as repressors. Both compete for binding to RORE elements in the BMAL1 promoter, exerting opposing effects on BMAL1 expression [24].
Additionally, basic helix-loop-helix proteins called differentiated embryo chondrocytes (DECs) are also CLOCK-BMAL1 targets. In a negative feedback loop, DECs inhibit CLOCK-BMAL1 activity by competing for E-box binding [25] (Figure 1).
BMAL1 can alternatively heterodimerize with neuronal PAS domain protein-2 (NPAS2), broadening the range of target genes under circadian regulation [20].
Another key participant in circadian rhythmicity is TIMELESS, which interacts with PER and CRY proteins to promote their nuclear localization and stability, thereby enhancing their repression of CLOCK-BMAL1 activity [26,27].
Circadian regulation also occurs during post-translational modifications. For example, (1) MAPK-dependent phosphorylation of BMAL1 at threonine 534 induces a conformational change that reduces its ability to bind E-box elements, inhibiting its transcriptional activity [28]; (2) SUMOylation of BMAL1 at lysine 259 is circadian and enhances BMAL1 activity [29]; (3) Acetylation of BMAL1 by CLOCK also increases its activity [30], whereas SIRT1-dependent deacetylation decreases it [31].

3. Possible Mechanisms Linking Circadian Disruption and HCC

Up to 40% of the transcriptome is under circadian control [32], regulating multiple functions essential for maintaining homeostasis (such as metabolism, immune responses, cell cycle progression, and cell death), which, when disrupted, may promote cancer.
Mouse models harboring mutant circadian genes exhibit an increased risk of cancer development. In these models, HCC is the second most frequent cancer, following lymphomas [33].
Mouse models exposed to chronic jet leg develop persistent metabolic derangements that lead to steatohepatitis and spontaneous HCC [34,35]. A central mechanism driving hepatic steatogenesis, fibrogenesis, and carcinogenesis in this model was the activation of the nuclear receptor constitutive androstane receptor (CAR), mediated by sympathetic dysfunction and cholestasis. Chronic jet leg disrupts the synchronization between the central and peripheral clocks via the sympathetic nervous system. During the sleep phase, sympathetic tone increases, inducing beta-adrenergic signaling, which activates protein kinase A (PKA)-dependent CAR signaling and MAPK-dependent AP-1 activation. AP-1 then upregulates c-Myc, promoting cell growth and survival [33]. Chronic jet leg also induces genome-wide metabolic reprogramming, altering cholesterol and bile acids metabolism, and ultimately causing intrahepatic cholestasis. Elevated bile acids levels further activate CAR. CAR is a potent driver of carcinogenesis through survival and proliferation pathways, including Wnt/beta-catenin signaling [34], which is one of the earliest and most important genetic alterations in human HCC [36]. Wnt/beta-catenin signaling is associated with HCC stemness, tumor growth, metastasis, and therapy resistance [36].
Another key pathway in HCC development is the Hedgehog pathway [37]. Hedgehog signaling in hepatocytes is under circadian control and, in turn, exerts negative feedback on the circadian clock by promoting the expression of the repressors PERs [38]. Experimental BMAL1 knockdown increases Hedgehog pathway activation. Notably, most HCC exhibit Hedgehog activation, which correlates with tumor growth, invasiveness, metastasis, and treatment resistance [39].
Circadian disruption can also blunt PER1-mediated activation of ATM and alter CLOCK-BMAL1-dependent SIRT1-mediated acetylation of p53. Loss of ATM and p53 activity results in defective apoptosis, impaired cell cycle arrest, and reduced DNA repair, leading to increased tumor growth and genomic instability [40].
The PI3K/AKT/mTOR signaling pathway is also under circadian regulation [41], which in turn modulates circadian clock function, including through CLOCK serine 845 phosphorylation, which decreases its nuclear localization [41] and through mechanisms required for CLOCK-BMAL1 heterodimerization and DNA binding [42]. This pathway plays a central oncogenic role in HCC, driving cell proliferation and survival and inducing metabolic reprogramming toward the Warburg effect (i.e., glycolysis to lactate production even in the presence of oxygen) [43]. In addition, this pathway promotes epithelial–mesenchymal transition and upregulates matrix metalloproteinases, thereby enhancing invasiveness and metastasis [44,45].
Upregulation of NPAS2, an alternative heterodimer to the BMAL1 partner, in some HCC tumors is associated with enhanced expression of CDC25A, which dephosphorylates cyclin-dependent kinases, promoting cell proliferation and inhibiting mitochondria-dependent intrinsic apoptosis. NPAS2 upregulation is associated with worse recurrence-free and overall survival [46].
In conclusion, the circadian pathway intersects with several carcinogenic pathways that play key roles in hepatocarcinogenesis. Indeed, HCC tumors presenting molecular signatures of circadian dysfunction activate pro-carcinogenic pathways, including Wnt/β-catenin, Hedgehog, and PI3K/ATK/mTOR, and inhibit tumor suppressors such as ATM and p53 [36,47,48] (Figure 2).

4. Epidemiological Evidence Linking Circadian Disruption and HCC

Circadian misalignment seems to be associated with an increased risk of HCC, although some conflicting reports highlight the need for further research on this topic. Population-level epidemiological studies evaluating HCC incidence across geographical longitudes within the same time zone have consistently shown an increased HCC risk from east to west [49,50,51], after adjustment for confounding factors such as the prevalence of health conditions (including obesity and diabetes mellitus), lifestyle, shift work occupation, socioeconomic status, and environmental factors. Each degree of longitude westward corresponds to a 4 min delay in sunrise [49]. Cities located on the western edges of a time zone may therefore experience more than 60 min of circadian misalignment compared with those on the eastern edge, resulting in a phase delay characterized by reduced morning light exposure and increased light exposure later in the day [52]. In contrast, a prospective cohort study including nearly half a million individuals did not observe an association between residential outdoor light-at-night exposure and HCC risk [53]. However, outdoor light-at-night exposure is difficult to quantify accurately, which may have led to an underestimation of its effect. Furthermore, although meta-analyses support an association between night-shift work and increased cancer-related mortality [54], the largest epidemiological study specifically assessing HCC unexpectedly reported a decreased liver cancer risk among Japanese men engaging in rotating shift work [55]. Of note, the Japanese study did not account for variations in shift work patterns, duration, intensity, or underlying HCC etiology. Moreover, the cohort—aged 40–79—may reflect a healthy-worker effect, as older individuals are less likely to engage in night shifts and those who remain cancer-free by age 79 are unlikely to experience substantial additional risk from continued exposure.
Sleep patterns appear to modulate the risk of HCC. Similar to what has been described for MASLD, cardiovascular diseases, and overall mortality [56], there seems to be a U-shaped association between sleep duration and HCC risk, with a nadir between 6 and 8 h of sleep [57,58], after adjustment for socioeconomic status, lifestyle habits, body mass index, and comorbidities, including diabetes mellitus and hypertension. Large epidemiological studies suggest that sleeping 5 h or less per day increases the risk of HCC by approximately 25%, whereas sleeping 9 h or more increases the risk by 25% to up to 2-fold (and up to 3-fold in obese patients) [57,58,59]. Daytime napping, particularly for more than one hour, which disrupts the circadian cycle, has also been associated with an almost 50% increased risk of HCC [58]. In addition to sleep quantity, sleep quality also appears to play an important role, with large epidemiological studies suggesting an almost 50% increase in HCC risk among patients with poor sleep quality [8,60]. It has further been estimated that nearly 15% of liver cancers could be prevented if the population maintained healthy sleep patterns [60]. It is important to note that epidemiological studies may be affected by reverse causality. In particular, some liver diseases, including liver cirrhosis, a premalignant condition that can progress to HCC, have been linked to sleep disorders, such as increased sleep fragmentation, reduced sleep efficiency, and longer daytime naps [56]. However, several design features mitigated the risk of reverse causation: all studies employed prospective cohort designs; three excluded participants with prior history of malignancy [8,58,59]; three excluded those with pre-existing chronic liver disease [57,59,60], with one additionally excluding individuals with hepatitis B or C infection [58]; and two excluded cases occurring within the first 2–5 years of follow up [58,60]. A summary of the epidemiological studies is presented in Table 1.
Human HCC frequently exhibits disturbances in circadian gene expression. Studies comparing gene expression in HCC tumors and adjacent non-tumor tissues have consistently shown decreased expression of several circadian genes in tumors, including PER1-3, CYR2, TIM, and NPAS2 [46,61,62,63,64,65,66,67,68]. In addition to reduced expression, some core clock components, such as PER1 and CRY1, lose their circadian oscillation in HCC samples [69]. Interestingly, these changes in gene expression are rarely associated with gene mutations [66]; instead, epigenetic modifications are the primary drivers of these dysregulations. For example, promoter methylation often silences these genes [61]. In HCC tumors, histone methyltransferases, such as EZH2, are overexpressed [61]. EZH2 itself exhibits circadian oscillation in expression [70] and can epigenetically modify clock genes, including members of the PER and CRY families [71]. Another epigenetic mechanism in HCC involves the overexpression of histone deacetylases (HDACs) [72,73], which promote chromatin condensation and reduce the accessibility of the transcriptional machinery to the promoters of core clock genes, such as BMAL1 and members of the PER and CRY families [74,75]. Non-coding RNAs may also contribute to circadian gene dysregulation in HCC. For instance, the expression of miR-195-5p and miR-122-5p is decreased in HCC samples, which appears to indirectly reduce the expression of circadian genes [76,77]. Conversely, the long non-coding RNA HULC is upregulated in HCC and increases CLOCK expression while altering its periodicity through complementary base pairing with the untranslated region of CLOCK mRNA [78].
These diverse mechanisms of circadian gene dysregulation underscore their fundamental role in HCC pathology, with significant prognostic implications. Different gene signatures, reflecting combinations of differentially expressed circadian genes in tumor and non-tumor tissues, have shown good accuracy in predicting prognosis. High-risk signatures, characterized by greater disruption of circadian rhythms, are associated with aggressive malignant highly proliferative phenotypes, higher pathological grade, advanced staging, and poorer overall survival [47,48,67,79,80,81,82,83,84]. They are also correlated with elevated expression of immune checkpoint genes, increased immune infiltration, a less immunocompetent phenotype, and reduced responsiveness to immunotherapy [67,82,84].
In addition to altered gene expression, germinal genetic variants of clock genes are critical factors linked to the prognosis of HCC patients. For instance, single nucleotide polymorphisms (SNPs) in PER1, PER3, CRY1, and NPAS2 have been associated with recurrence-free and overall survival following surgical resection [85] and transarterial chemoembolization [86,87]. A dose-dependent effect was observed, with a higher number of variants corresponding to a stronger impact on prognosis.

5. Chronotherapy for HCC

The term chronotherapy encompasses two distinct concepts: (1) interventions that align the circadian rhythm and (2) the scheduling of therapeutic interventions at specific times of day [52,88].
One way to entrain the circadian rhythm is by manipulating feeding schedules. In mouse models of HCC, time-restricting eating (TRE) reduces both the development and growth of liver tumors [89,90] and is associated with higher tumor differentiation and lower metastatic potential [91]. The antitumor effects of TRE may stem from circadian resynchronization, which occurs through several mechanisms: (1) fasting decreases CRY1 and PER proteins stability via AMPK-dependent pathways, enabling the restoration of clock genes oscillation [22,92]; (2) fasting increases SIRT1 activity, which interacts with CLOCK-BMAL1 and suppresses PER2 transcription [93]; and (3) feeding induces CRY gene expression in an mTOR-dependent but CLOCK-BMAL1 independent manner [94]. In addition, prolonged fasting may exert antitumor effects, as tumor cells are more vulnerable to nutrient depletion. Their higher susceptibility to fasting arises from a shift toward less energetically efficient anaerobic glucose metabolism, inability to downregulate cell proliferation and energy demands, and lack of compensatory autophagy under nutrient-depleted conditions [90,95]. Forcing tumor cells to shift from anaerobic metabolism to oxidative phosphorylation increases oxidative stress, DNA damage, and apoptosis in cells that maintain a high proliferative index [90]. Animal models have also shown that intermittent fasting, but not short-term fasting, modifies the tumor microenvironment by altering the extracellular matrix, vascularization, and increasing immune cell infiltration. These changes enhance antitumor drug delivery and affect cancer cell survival, invasion, and metastasis [90]. Preclinical in vitro and in vivo studies have suggested that fasting may increase the response to sorafenib treatment. One explanation could be sorafenib’s inhibition of mitochondrial respiration, which increases cellular vulnerability to nutrient depletion [95]. No clinical trials have yet evaluated TRE in human HCC, and caution is warranted regarding the potential risks of TRE in cirrhotic patients with HCC, as fasting increases the risk of sarcopenia in patients with cirrhosis [96].
Melatonin is produced in the pineal gland in coordination with the light–dark circadian cycle and conveys this information to regulate sleep time [97]. In addition to its chronobiological role, melatonin possesses antioxidant, anti-inflammatory, and antiproliferative properties [98,99], which contribute to its intrinsic antitumor activity in various cancers, including HCC [97]. In vitro studies using human HCC cell lines have shown that melatonin induces apoptosis and inhibits cell proliferation, epithelial–mesenchymal transition, migration, and invasiveness [100]. Melatonin also exhibits anti-angiogenic properties [101]. These effects are mediated through multiple mechanisms, including FOXA-2- and mTORC1-dependent inhibition of hypoxia-inducible factor-1 (HIF-1) [97,102], modulation of cAMP, ERK/MAPK-JNK, and MAPK-P38 signaling [103,104], NF-kB inhibition-dependent upregulation of tissue inhibitor of metalloproteinases-1 (TIMP-1) [105], and metabolic reprograming that counteracts the Warburg effect, by which cancer cells ferment glucose to lactate despite adequate oxygen availability [106]. In vivo HCC models have similarly demonstrated that melatonin administration abrogates tumor growth [97]. Notably, similar tumor growth inhibition was observed when animals were exposed to light-emitting diode illumination, which amplified nocturnal melatonin levels compared to cool white fluorescent lighting [106]. Melatonin treatment ultimately improved survival in these HCC models [107]. In vitro data also suggest that melatonin increases HCC cell sensitivity to sorafenib treatment [102,108]. It might even modulate immunotherapy responses, as exosomes from melatonin-treated HCC cells promote a more immunocompetent phenotype and suppress PD-L1 expression in vitro and in vivo [104,109]. A comprehensive analysis of 12 melatoninergic genes showed that tumor tissues exhibit reduced expression of nuclear and cytoplasmic melatonin receptor genes, as well as metabolic melatonin genes, compared to adjacent non-tumor tissues [110]. Lower tumor expression of melatonin-synthesizing enzymes has also been linked to increased mortality in patients with HCC [111]. Furthermore, among patients with HCC undergoing liver transplantation, lower pre-transplant melatonin levels were associated with a higher risk of 1-year post-transplantation mortality [112]. Despite the growing evidence supporting the rationale for melatonin as a therapeutic strategy for HCC, no clinical trials have evaluated melatonin treatment in human HCC.
Regarding the optimization of treatment timing, a seminal paper published nearly 30 years ago on colorectal cancer first reported that timed infusions of chemotherapy were associated with an increased response rate and reduced toxicity, allowing the administration of higher doses of fluorouracil [113]. Since then, several studies on different cancers have evaluated the effect of the timing of chemotherapy administration, with the majority describing decreased toxicity and some describing increased efficacy [114]. Prespecified time-of-day treatment delivery relies on circadian clock effects in different aspects of drug delivery, including absorption (by modulating gastric emptying, gut motility and blood flow, drug transporter and exporter expression [115,116,117]), distribution (by differences in cardiac output and blood flow across the day [118]), metabolism (for example, clock genes regulate the expression of cytochrome P450 enzymes [119,120,121,122]), and excretion [52].
HCC is insensitive to chemotherapy; however, in vitro and in vivo preclinical models suggest that the timing of the administration of the tyrosine kinase inhibitor sorafenib has an impact on tumor progression [123]. Data on the time-of-day administration of tyrosine kinase inhibitors in human HCC are still lacking.
HCC preclinical models have shown a therapeutic advantage and lower toxicity when radiation therapy was administered during the early activity phase or late inactivity phase, which corresponded to peaks of HCC cell proliferation [124,125]. In addition, adjusting radiation timing to circadian fluctuations in DNA repair machinery may impact treatment-related toxicity [88]. The effect of radiation timing on human HCC remains undetermined.
In the last five years, the establishment of immunotherapy for HCC treatment has revolutionized systemic therapy. Tumor cells can evade immune attack by expressing programmed death-ligand-1 (PD-L1), which binds to its corresponding receptor on immune cells, programmed cell death protein-1 (PD-1), thereby inhibiting immune cell activity. Additionally, cytotoxic T-lymphocyte antigen-4 (CTLA-4), which is expressed on T cells, suppresses T-cell responses by competing with the costimulatory receptor CD28, which is required for complete T-cell activation. Immune checkpoint inhibitors (ICI) are monoclonal antibodies that block PD-L1, PD-1, or CTLA-4, preventing their inhibitory effects and eliciting an antitumor immune response [126]. Some patients achieve long-term survival [127] but fewer than one-third experience an objective response [128]. Chronotherapy is one strategy to improve response rates. Retrospective studies on several metastatic cancers have suggested that early day administration of immunotherapy is associated with increased response rates and improved overall survival [126,129]. In HCC, preclinical models suggested that circadian-timed administration of immunotherapy, compared with continuous infusion, led to a 4-fold increase in the objective response rate and eliminated mortality [130]. More recently, a retrospective analysis of human HCC, including a total of 751 patients, reported that morning infusions of ICIs, compared with afternoon infusions, were associated with improved progression-free and overall survival and a higher objective response rate (45% vs. 33%) [131]. Despite providing valuable insights, this study has several limitations inherent to its retrospective design. First, unmeasured confounding factors may have influenced the observed associations, including variables such as liver function fluctuations (even though the study only included patients from Child-Pugh-Turcotte class A at baseline), comorbidities, prior therapies, tumor molecular profile, and adherence to treatment protocols. Second, selection bias cannot be entirely excluded, as patients included in retrospective cohorts may differ systematically from those not captured in the dataset. Third, timing and accuracy of clinical and laboratory measurements may vary, potentially affecting the assessment of predictors and outcomes. These limitations underscore the need for well-designed prospective studies to rigorously evaluate predictors of immunotherapy response in hepatocellular carcinoma. Although such trials are currently ongoing for melanoma (NCT07155317) and lung cancer (NCT05549037), comparable studies specifically addressing HCC are still lacking. The effects of immunotherapy timing on pharmacokinetics would be expected to manifest primarily before the drug reaches steady-state blood concentrations, which occurs between the second and fourth months of therapy [132], and hence the timing of the first infusions would be particularly relevant [126]. However, blood levels do not accurately reflect antibody levels within the tumor [133], as uptake is influenced by competition between tumor-infiltrating immune cells and lymphoid organs, which may exhibit circadian oscillations [134]. The immune response appears to be subject to circadian regulation. The number and function of tumor-infiltrating T cells vary throughout the day [135]. Several mechanisms have been proposed, including the regulation of T cell infiltration by endothelial circadian clocks, which drive oscillations in endothelial cell adhesion molecule expression [136]. The phenotype of tumor-associated immune cells is also under circadian control, with oscillating genes enriched in pathways related to metabolism, cell adhesion, differentiation, and T cell activation [137]. Notably, PD-1 expression in tumor-infiltrating T cells also follows a circadian pattern [135].

6. Conclusions

HCC is a deadly cancer with a rising incidence, particularly in Western countries, largely driven by steatotic liver diseases. Notably, more than 50% of the attributable risk is related to lifestyle factors, meaning that up to half of all HCC cases can be prevented. Modern lifestyles increasingly disregard circadian rhythms, with artificial light altering natural light–dark cycles and food being available around the clock. Disruption of the circadian cycle not only contributes to liver diseases that can progress to HCC but also appears to have intrinsic hepato-oncogenic effects.
The most effective way to reduce HCC mortality is by reducing its incidence. Strong educational efforts and government policies should be implemented to promote lifestyles that align with healthy circadian rhythms.
Systemic treatment of HCC has dramatically changed in the last five years with the advent of immunotherapy. However, only approximately one-third of patients respond to these treatments. Chronotherapy is gaining attention and appears to be a promising strategy for improving therapeutic responses. Assessing tumor circadian disruption may help identify patients who are most likely to benefit from the treatment.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
BMAL1Brain and Muscle ARNT-Like-1
CARConstitutive Androstane Receptor
CLOCKCircadian Locomotor Output Cycle Kaput
CRYCryptochrome
CTLA-4Cytotoxic T-Lymphocyte Antigen-4
DECDifferentiated Embryo Chondrocytes
HCCHepatoCellular Carcinoma
HDACHistone DdeACetylase
IARCInternational Agency for Research on Cancer
ICIImmune Checkpoint Inhibitors
MARCKSL1Myristoylated Alanine-Rich C Kinase Substrate-Like 1
MASLDMetabolic dysfunction-Associated Steatotic Liver Disease
NNMTNicotinamide N-MethylTransferase
NPAS2Neuronal PAS domain Protein-2
PD-1Programmed cell Death protein-1
PD-L1Programmed Death-Ligand-1
PERPeriod
PKAProtein Kinase A
RORRetinoic acid-related Orphan nuclear Receptors
SLDSteatotic Liver Disease
SNPSingle Nucleotide Polymorphisms
TIMP-1Tissue Inhibitor of MetalloProteinases-1
TRETime Restricting Eating

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Vogel, A.; Meyer, T.; Sapisochin, G.; Salem, R.; Saborowski, A. Hepatocellular carcinoma. Lancet 2022, 400, 1345–1362. [Google Scholar] [CrossRef]
  3. Rumgay, H.; Arnold, M.; Ferlay, J.; Lesi, O.; Cabasag, C.J.; Vignat, J.; Laversanne, M.; McGlyn, K.A.; Soeromataram, I. Global burden of primary liver cancer in 2020 and predictions to 2040. J. Hepatol. 2022, 77, 1598–1606. [Google Scholar] [CrossRef]
  4. Adhoute, X.; Penaranda, G.; Raoul, J.L.; Edeline, J.; Blanc, J.F.; Pol, B.; Campanile, M.; Perrier, H.; Bayle, O.; Monnet, O.; et al. Barcelona clinic liver cancer nomogram and others staging/scoring systems in a french hepatocellular carcinoma cohort. World J. Gastroenterol. 2017, 23, 2545–2555. [Google Scholar] [CrossRef]
  5. Vitale, A.; Morales, R.R.; Zanus, G.; Farinati, F.; Burra, P.; Angeli, P.; Frigo, A.C.; Del Poggio, P.; Rapaccini, G.; Di Nolfo, M.A.; et al. Barcelona clinic liver cancer staging and transplant survival benefit for patients with hepatocellular carcinoma: A multicentre, cohort study. Lancet Oncol. 2011, 12, 654–662. [Google Scholar] [CrossRef]
  6. Huang, D.Q.; Singal, A.G.; Kono, Y.; Tan, D.J.H.; El-Serag, H.B.; Loomba, R. Changing global epidemiology of liver cancer from 2010 to 2019: Nash is the fastest growing cause of liver cancer. Cell Metab. 2022, 34, 969–977.e2. [Google Scholar] [CrossRef]
  7. Zhou, K.; Lim, T.; Dodge, J.L.; Terrault, N.A.; Wilkens, L.R.; Setiawan, V.W. Population-attributable risk of modifiable lifestyle factors to hepatocellular carcinoma: The multi-ethnic cohort. Aliment. Pharmacol. Ther. 2023, 58, 89–98. [Google Scholar] [CrossRef] [PubMed]
  8. Hu, L.Y.; Chen, P.M.; Hu, Y.W.; Shen, C.C.; Perng, C.L.; Su, T.P.; Yen, S.H.; Tzeng, C.H.; Chiou, T.J.; Yeh, C.M.; et al. The risk of cancer among patients with sleep disturbance: A nationwide retrospective study in taiwan. Ann. Epidemiol. 2013, 23, 757–761. [Google Scholar] [CrossRef] [PubMed]
  9. Erren, T.C.; Morfeld, P. Circadian epidemiology: Structuring circadian causes of disease and practical implications. Chronobiol. Int. 2024, 41, 38–52. [Google Scholar] [CrossRef]
  10. Verdelho Machado, M. Circadian deregulation: Back facing the sun toward metabolic dysfunction-associated steatotic liver disease (MASLD) development. Nutrients 2024, 16, 4294. [Google Scholar] [CrossRef] [PubMed]
  11. Mazzoccoli, G.; Miele, L.; Marrone, G.; Mazza, T.; Vinciguerra, M.; Grieco, A. A role for the biological clock in liver cancer. Cancers 2019, 11, 1778. [Google Scholar] [CrossRef]
  12. Hurni, C.; Weger, B.D.; Gobet, C.; Naef, F. Comprehensive analysis of the circadian nuclear and cytoplasmic transcriptome in mouse liver. PLoS Genet. 2022, 18, e1009903. [Google Scholar] [CrossRef]
  13. Reinke, H.; Asher, G. Circadian clock control of liver metabolic functions. Gastroenterology 2016, 150, 574–580. [Google Scholar] [CrossRef]
  14. Stokkan, K.A.; Yamazaki, S.; Tei, H.; Sakaki, Y.; Menaker, M. Entrainment of the circadian clock in the liver by feeding. Science 2001, 291, 490–493. [Google Scholar] [CrossRef] [PubMed]
  15. Ko, C.H.; Takahashi, J.S. Molecular components of the mammalian circadian clock. Hum. Mol. Genet. 2006, 15, R271–R277. [Google Scholar] [CrossRef] [PubMed]
  16. Gekakis, N.; Staknis, D.; Nguyen, H.B.; Davis, F.C.; Wilsbacher, L.D.; King, D.P.; Takahashi, J.S.; Weitz, C.J. Role of the clock protein in the mammalian circadian mechanism. Science 1998, 280, 1564–1569. [Google Scholar] [CrossRef]
  17. Yan, J.; Wang, H.; Liu, Y.; Shao, C. Analysis of gene regulatory networks in the mammalian circadian rhythm. PLoS Comput. Biol. 2008, 4, e1000193. [Google Scholar] [CrossRef] [PubMed]
  18. Zheng, B.; Albrecht, U.; Kaasik, K.; Sage, M.; Lu, W.; Vaishnav, S.; Li, K.; Sun, Z.S.; Eichele, G.; Bradley, A.; et al. Nonredundant roles of the mper1 and mper2 genes in the mammalian circadian clock. Cell 2001, 105, 683–694. [Google Scholar] [CrossRef]
  19. Kume, K.; Zylka, M.J.; Sriram, S.; Shearman, L.P.; Weaver, D.R.; Jin, X.; Maywood, E.S.; Hastings, M.H.; Reppert, S.M. Mcry1 and mcry2 are essential components of the negative limb of the circadian clock feedback loop. Cell 1999, 98, 193–205. [Google Scholar] [CrossRef]
  20. Crespo, M.; Leiva, M.; Sabio, G. Circadian clock and liver cancer. Cancers 2021, 13, 3631. [Google Scholar] [CrossRef]
  21. Knippschild, U.; Gocht, A.; Wolff, S.; Huber, N.; Lohler, J.; Stoter, M. The casein kinase 1 family: Participation in multiple cellular processes in eukaryotes. Cell Signal 2005, 17, 675–689. [Google Scholar] [CrossRef] [PubMed]
  22. Lamia, K.A.; Sachdeva, U.M.; DiTacchio, L.; Williams, E.C.; Alvarez, J.G.; Egan, D.F.; Vasquez, D.S.; Juguilon, H.; Panda, S.; Shaw, R.J.; et al. AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science 2009, 326, 437–440. [Google Scholar] [CrossRef] [PubMed]
  23. Correia, S.P.; Chan, A.B.; Vaughan, M.; Zolboot, N.; Perea, V.; Huber, A.L.; Kriebs, A.; Moresco, J.J.; Yates, R., 3rd; Lamia, K.A. The circadian e3 ligase complex scf(fbxl3+cry) targets tlk2. Sci. Rep. 2019, 9, 198. [Google Scholar] [CrossRef]
  24. Guillaumond, F.; Dardente, H.; Giguere, V.; Cermakian, N. Differential control of bmal1 circadian transcription by rev-erb and ror nuclear receptors. J. Biol. Rhythms 2005, 20, 391–403. [Google Scholar] [CrossRef] [PubMed]
  25. Honma, S.; Kawamoto, T.; Takagi, Y.; Fujimoto, K.; Sato, F.; Noshiro, M.; Kato, Y.; Honma, K.I. Dec1 and dec2 are regulators of the mammalian molecular clock. Nature 2002, 419, 841–844. [Google Scholar] [CrossRef]
  26. Barnes, J.W.; Tischkau, S.A.; Barnes, J.A.; Mitchell, J.W.; Burgoon, P.W.; Hickok, J.R.; Gillette, M.U. Requirement of mammalian timeless for circadian rhythmicity. Science 2003, 302, 439–442. [Google Scholar] [CrossRef]
  27. Engelen, E.; Janssens, R.C.; Yagita, K.; Smits, V.A.; van der Horst, G.T.; Tamanini, F. Mammalian timeless is involved in period determination and DNA damage-dependent phase advancing of the circadian clock. PLoS ONE 2013, 8, e56623. [Google Scholar] [CrossRef]
  28. Sanada, K.; Okano, T.; Fukada, Y. Mitogen-activated protein kinase phosphorylates and negatively regulates basic helix-loop-helix-pas transcription factor bmal1. J. Biol. Chem. 2002, 277, 267–271. [Google Scholar] [CrossRef]
  29. Cardone, L.; Hirayama, J.; Giordano, F.; Tamaru, T.; Palvimo, J.J.; Sassone-Corsi, P. Circadian clock control by sumoylation of bmal1. Science 2005, 309, 1390–1394. [Google Scholar] [CrossRef]
  30. Hirayama, J.; Sahar, S.; Grimaldi, B.; Tamaru, T.; Takamatsu, K.; Nakahata, Y.; Sassone-Corsi, P. Clock-mediated acetylation of bmal1 controls circadian function. Nature 2007, 450, 1086–1090. [Google Scholar] [CrossRef]
  31. Nakahata, Y.; Kaluzova, M.; Grimaldi, B.; Sahar, S.; Hirayama, J.; Chen, D.; Guarente, L.P.; Sassoni-Corsi, P. The nad+-dependent deacetylase sirt1 modulates clock-mediated chromatin remodeling and circadian control. Cell 2008, 134, 329–340. [Google Scholar] [CrossRef]
  32. Zhang, R.; Lahens, N.F.; Ballance, H.I.; Hughes, M.E.; Hogenesch, J.B. A circadian gene expression atlas in mammals: Implications for biology and medicine. Proc. Natl. Acad. Sci. USA 2014, 111, 16219–16224. [Google Scholar] [CrossRef]
  33. Lee, S.; Donehower, L.A.; Herron, A.J.; Moore, D.D.; Fu, L. Disrupting circadian homeostasis of sympathetic signaling promotes tumor development in mice. PLoS ONE 2010, 5, e10995. [Google Scholar] [CrossRef] [PubMed]
  34. Kettner, N.M.; Voicu, H.; Finegold, M.J.; Coarfa, C.; Sreekumar, A.; Putluri, N.; Katchy, C.A.; Lee, C.; Moore, D.D.; Fu, L. Circadian homeostasis of liver metabolism suppresses hepatocarcinogenesis. Cancer Cell 2016, 30, 909–924. [Google Scholar] [CrossRef]
  35. Padilla, J.; Osman, N.M.; Bissig-Choisat, B.; Grimm, S.L.; Qin, X.; Major, A.M.; Yang, L.; Lopez-Terrada, D.; Coarfa, C.; Li, F. Circadian dysfunction induces nafld-related human liver cancer in a mouse model. J. Hepatol. 2024, 80, 282–292. [Google Scholar]
  36. Xu, C.; Xu, Z.; Zhang, Y.; Evert, M.; Calvisi, D.F.; Chen, X. Beta-catenin signaling in hepatocellular carcinoma. J. Clin. Investig. 2022, 132, e154515. [Google Scholar] [CrossRef]
  37. Ding, J.; Li, H.Y.; Zhang, L.; Zhou, Y.; Wu, J. Hedgehog signaling, a critical pathway governing the development and progression of hepatocellular carcinoma. Cells 2021, 10, 123. [Google Scholar] [CrossRef]
  38. Marbach-Breitruck, E.; Matz-Soja, M.; Abraham, U.; Schmidt-Heck, W.; Sales, S.; Rennert, C.; Kern, M.; Aleithe, S.; Spormann, L.; Thiel, C. Tick-tock hedgehog-mutual crosstalk with liver circadian clock promotes liver steatosis. J. Hepatol. 2019, 70, 1192–1202. [Google Scholar] [CrossRef] [PubMed]
  39. Jeng, K.S.; Chang, C.F.; Tsang, Y.M.; Sheen, I.S.; Jeng, C.J. Reappraisal of the roles of the sonic hedgehog signaling pathway in hepatocellular carcinoma. Cancers 2024, 16, 1739. [Google Scholar] [CrossRef] [PubMed]
  40. Mteyrek, A.; Filipski, E.; Guettier, C.; Okyar, A.; Levi, F. Clock gene per2 as a controller of liver carcinogenesis. Oncotarget 2016, 7, 85832–85847. [Google Scholar] [CrossRef] [PubMed]
  41. Luciano, A.K.; Zhou, W.; Santana, J.M.; Kyriakides, C.; Velazquez, H.; Sessa, W.C. Clock phosphorylation by akt regulates its nuclear accumulation and circadian gene expression in peripheral tissues. J. Biol. Chem. 2018, 293, 9126–9136. [Google Scholar] [CrossRef]
  42. Morishita, Y.; Miura, D.; Kida, S. Pi3k regulates bmal1/clock-mediated circadian transcription from the dbp promoter. Biosci. Biotechnol. Biochem. 2016, 80, 1131–1140. [Google Scholar] [CrossRef]
  43. Paskeh, M.D.A.; Ghadyani, F.; Hashemi, M.; Abbaspour, A.; Zabolian, A.; Javanshir, S.; Razzazan, M.; Mirzaei, S.; Entezari, M.; Goharrizi, M.; et al. Biological impact and therapeutic perspective of targeting pi3k/akt signaling in HCC: Promises and challenges. Pharmacol. Res. 2023, 187, 106553. [Google Scholar] [CrossRef]
  44. Larue, L.; Bellacosa, A. Epithelial-mesenchymal transition in development and cancer: Role of phosphatidylinositol 3′ kinase/akt pathways. Oncogene 2005, 24, 7443–7454. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, J.S.; Wang, Q.; Fu, X.H.; Huang, X.H.; Chen, X.L.; Cao, L.Q.; Chen, L.Z.; Tan, H.X.; Li, W.; Bi, J.; et al. Involvement of pi3k/pten/akt/mtor pathway in invasion and metastasis in hepatocellular carcinoma: Association with mmp-9. Hepatol. Res. 2009, 39, 177–186. [Google Scholar] [CrossRef] [PubMed]
  46. Yuan, P.; Li, J.; Zhou, F.; Huang, Q.; Zhang, J.; Guo, X.; Lyu, Z.; Zhang, H.; Xing, J. NPAS2 promotes cell survival of hepatocellular carcinoma by transactivating CDC25a. Cell Death Dis. 2017, 8, e2704. [Google Scholar] [CrossRef]
  47. Liang, Y.; Wang, S.; Huang, X.; Chai, R.; Tang, Q.; Yang, R.; Huang, X.; Wang, X.; Zheng, K. Dysregulation of circadian clock genes as significant clinic factor in the tumorigenesis of hepatocellular carcinoma. Comput. Math. Methods Med. 2021, 2021, 8238833. [Google Scholar] [CrossRef]
  48. Wu, Z.; Hu, H.; Zhang, Q.; Wang, T.; Li, H.; Qin, Y.; Ai, X.; Yi, W.; Wei, X.; Gao, W.; et al. Four circadian rhythm-related genes predict incidence and prognosis in hepatocellular carcinoma. Front. Oncol. 2022, 12, 937403. [Google Scholar] [CrossRef]
  49. VoPham, T.; Weaver, M.D.; Vetter, C.; Hart, J.E.; Tamimi, R.M.; Laden, F.; Bertrand, K.A. Circadian misalignment and hepatocellular carcinoma incidence in the united states. Cancer Epidemiol. Biomark. Prev. 2018, 27, 719–727. [Google Scholar] [CrossRef]
  50. Gu, F.; Xu, S.; Devesa, S.S.; Zhang, F.; Klerman, E.B.; Graubard, B.I.; Caporaso, N.E. Longitude position in a time zone and cancer risk in the united states. Cancer Epidemiol. Biomark. Prev. 2017, 26, 1306–1311. [Google Scholar] [CrossRef] [PubMed]
  51. Niu, J.; Brown, C.; Law, M.; Colacino, J.A.; Ritov, Y. Longitudinal position and cancer risk in the united states revisited. Cancer Res. Commun. 2024, 4, 328–336. [Google Scholar] [CrossRef]
  52. Ferrell, J.M. Chronobiology of cancers in the liver and gut. Cancers 2024, 16, 2925. [Google Scholar] [CrossRef] [PubMed]
  53. Park, Y.; Ramirez, Y.; Xiao, Q.; Liao, L.M.; Jones, G.S.; McGlynn, K.A. Outdoor light at night and risk of liver cancer in the nih-aarp diet and health study. Cancer Causes Control 2022, 33, 1215–1218. [Google Scholar] [CrossRef]
  54. Su, F.; Huang, D.; Wang, H.; Yang, Z. Associations of shift work and night work with risk of all-cause, cardiovascular and cancer mortality: A meta-analysis of cohort studies. Sleep Med. 2021, 86, 90–98. [Google Scholar] [CrossRef]
  55. Arafa, A.; Eshak, E.S.; Iso, H.; Muraki, I.; Tamakoshi, A. Night work, rotating shift work, and the risk of cancer in japanese men and women: The JACC study. J. Epidemiol. 2021, 31, 585–592. [Google Scholar] [CrossRef]
  56. Machado, M.V. The beauty sleep to keep a healthy liver. Int. J. Mol. Sci. 2025, 26, 11322. [Google Scholar] [CrossRef]
  57. Wang, Q.; Chen, H.; Deng, H.; Zhang, M.; Hu, H.; Ouyang, H.; Ma, L.; Liu, R.; Sun, J.; Hu, G.; et al. Association of daily sleep duration with risk of metabolic dysfunction-associated steatotic liver disease and adverse liver outcomes. Diabetes Metab. 2025, 51, 101628. [Google Scholar] [CrossRef]
  58. Long, L.; Zhao, L.; Petrick, J.L.; Liao, L.M.; Huang, T.; Hakim, A.; Yang, W.; Campbell, P.T.; Giovnnucci, E.; McGlyn, K.A.; et al. Daytime napping, nighttime sleeping duration, and risk of hepatocellular carcinoma and liver disease-related mortality. JHEP Rep. 2023, 5, 100819. [Google Scholar] [CrossRef]
  59. Royse, K.E.; El-Serag, H.B.; Chen, L.; White, D.L.; Hale, L.; Sangi-Haghpeykar, H.; Jiao, L. Sleep duration and risk of liver cancer in postmenopausal women: The women’s health initiative study. J. Womens Health 2017, 26, 1270–1277. [Google Scholar] [CrossRef] [PubMed]
  60. Wang, W.; Zhuang, Z.; Song, Z.; Zhao, Y.; Huang, T. Sleep patterns, genetic predisposition, and risk of chronic liver disease: A prospective study of 408,560 uk biobank participants. J. Affect. Disord. 2024, 352, 229–236. [Google Scholar] [CrossRef] [PubMed]
  61. Lin, Y.M.; Chang, J.H.; Yeh, K.T.; Yang, M.Y.; Liu, T.C.; Lin, S.F.; Su, W.W.; Chang, G. Disturbance of circadian gene expression in hepatocellular carcinoma. Mol. Carcinog. 2008, 47, 925–933. [Google Scholar] [CrossRef]
  62. Yang, S.L.; Yu, C.; Jiang, J.X.; Liu, L.P.; Fang, X.; Wu, C. Hepatitis b virus x protein disrupts the balance of the expression of circadian rhythm genes in hepatocellular carcinoma. Oncol. Lett. 2014, 8, 2715–2720. [Google Scholar] [CrossRef]
  63. Zhao, H.; Han, G.; Jiang, Z.; Gao, D.; Zhang, H.; Yang, L.; Ma, T.; Gao, L.; Wang, A.; Chao, W.; et al. Identification of bmal1-regulated circadian genes in mouse liver and their potential association with HCC: Gys2 and upp2 as promising candidates. Biochem. Biophys. Res. Commun. 2024, 696, 149422. [Google Scholar] [CrossRef]
  64. Chen, X.; Zhao, Q.; Wang, H.; Qiu, K.; Deng, X.; Xu, F. Period2 is associated with immune cell infiltration and is a potential diagnostic and prognostic marker for hepatocellular carcinoma. Front. Mol. Biosci. 2023, 10, 1264553. [Google Scholar] [CrossRef]
  65. Li, H.; Lu, Y.F.; Chen, H.; Liu, J. Dysregulation of metallothionein and circadian genes in human hepatocellular carcinoma. Chronobiol. Int. 2017, 34, 192–202. [Google Scholar]
  66. Liang, Q.; Ye, Y.; Li, E.; Fan, J.; Gong, J.; Ying, J.; Cao, Y.; Li, R.; Wang, P. A circadian clock gene-related signature for predicting prognosis and its association with sorafenib response in hepatocellular carcinoma. Transl. Cancer Res. 2023, 12, 2493–2507. [Google Scholar] [CrossRef]
  67. Ye, Z.; Du, Y.; Yu, W.; Lin, Y.; Zhang, L.; Chen, X. Construction of a circadian rhythm-relevant gene signature for hepatocellular carcinoma prognosis, immunotherapy and chemosensitivity prediction. Heliyon 2024, 10, e33682. [Google Scholar] [CrossRef]
  68. Huang, L.H.; Huang, C.Y.; Liu, Y.W.; Chien, P.C.; Hsieh, T.M.; Liu, H.T.; Lin, H.P.; Wu, C.J.; Chuang, P.C.; Hsieh, C.H. Circadian rhythm disruption in hepatocellular carcinoma investigated by integrated analysis of bulk and single-cell RNA sequencing data. Int. J. Mol. Sci. 2024, 25, 5748. [Google Scholar] [CrossRef]
  69. Anafi, R.C.; Francey, L.J.; Hogenesch, J.B.; Kim, J. Cyclops reveals human transcriptional rhythms in health and disease. Proc. Natl. Acad. Sci. USA 2017, 114, 5312–5317. [Google Scholar] [CrossRef]
  70. Zhong, Y.; Ye, Q.; Chen, C.; Wang, M.; Wang, H. Ezh2 promotes clock function and hematopoiesis independent of histone methyltransferase activity in zebrafish. Nucleic Acids Res. 2018, 46, 3382–3399. [Google Scholar] [CrossRef]
  71. Etchegaray, J.P.; Yang, X.; DeBruyne, J.P.; Peters, A.; Weaver, D.R.; Jenuwein, T.; Reppert, S.M. The polycomb group protein ezh2 is required for mammalian circadian clock function. J. Biol. Chem. 2006, 281, 21209–21215. [Google Scholar] [CrossRef]
  72. Freese, K.; Seitz, T.; Dietrich, P.; Lee, S.M.L.; Thasler, W.E.; Bosserhoff, A.; Hellerbrand, C. Histone deacetylase expressions in HCC and functional effects of histone deacetylase inhibitors on liver cancer cells in vitro. Cancers 2019, 11, 1587. [Google Scholar]
  73. Wang, J.; Liu, R.; Wang, Y.; Mo, H.; Niu, Y.; Xu, Q.; Liu, Q. Repression of the mir-627-5p by histone deacetylase 3 contributes to hypoxia-induced hepatocellular carcinoma progression. J. Cancer 2021, 12, 5320–5330. [Google Scholar]
  74. Fogg, P.C.; O’Neill, J.S.; Dobrzycki, T.; Calvert, S.; Lord, E.C.; McIntosh, R.L.; Elliott, C.J.H.; Sweeney, S.T.; Hastings, M.H.; Chawla, S. Class IIa histone deacetylases are conserved regulators of circadian function. J. Biol. Chem. 2014, 289, 34341–34348. [Google Scholar] [CrossRef]
  75. Sadia, K.; Castagna, A.; Udali, S.; Ambrosani, F.; Pattini, P.; Beri, R.; Argentino, G.; Masutti, S.; Riso, S. Epigenetic regulation through histone deacetylation: Implications and therapeutic potential in hepatocellular carcinoma. Cells 2025, 14, 1337. [Google Scholar] [CrossRef] [PubMed]
  76. Gu, Y.; Wei, X.; Sun, Y.; Gao, H.; Zheng, X.; Wong, L.L.; Jin, L.; Liu, N.; Hernandez, B.; Peplowska, K.; et al. Mir-192-5p silencing by genetic aberrations is a key event in hepatocellular carcinomas with cancer stem cell features. Cancer Res. 2019, 79, 941–953. [Google Scholar] [CrossRef] [PubMed]
  77. Wen, D.Y.; Huang, J.C.; Wang, J.Y.; Pan, W.Y.; Zeng, J.H.; Pang, Y.Y.; Yang, H. Potential clinical value and putative biological function of mir-122-5p in hepatocellular carcinoma: A comprehensive study using microarray and rna sequencing data. Oncol. Lett. 2018, 16, 6918–6929. [Google Scholar] [CrossRef]
  78. Cui, M.; Zheng, M.; Sun, B.; Wang, Y.; Ye, L.; Zhang, X. A long noncoding rna perturbs the circadian rhythm of hepatoma cells to facilitate hepatocarcinogenesis. Neoplasia 2015, 17, 79–88. [Google Scholar] [CrossRef]
  79. Wu, Y.; Shen, C.; Wang, X.; Zhao, W.; Li, Y.; He, X.; Chen, Y.; Liu, J.; Wu, X.; Shen, A. Development and validation of a novel circadian rhythm-related signature to predict the prognosis of the patients with hepatocellular carcinoma. Biomed. Res. Int. 2022, 2022, 4263261. [Google Scholar]
  80. Li, X.J.; Chang, L.; Mi, Y.; Zhang, G.; Zhu, S.S.; Zhang, Y.X.; Wang, H.Y.; Lu, Y.S.; Ping, Y.X.; Zheng, P.Y.; et al. Integrated-omics analysis defines subtypes of hepatocellular carcinoma based on circadian rhythm. J. Integr. Med. 2025, 23, 445–456. [Google Scholar] [CrossRef]
  81. Xu, Z.; Huang, W.; Zou, X.; Liu, S. Integrated machine learning constructed a circadian-rhythm-related model to assess clinical outcomes and therapeutic advantages in hepatocellular carcinoma. Transl. Cancer Res. 2025, 14, 1799–1823. [Google Scholar] [PubMed]
  82. Zhang, Z.; Gao, W.; Tan, X.; Deng, T.; Zhou, W.; Jian, H.; Zeng, P. Construction and verification of a novel circadian clock related long non-coding RNA model and prediction of treatment for survival prognosis in patients with hepatocellular carcinoma. BMC Cancer 2023, 23, 57. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, Z.; Liang, Z.; Gao, W.; Yu, S.; Hou, Z.; Li, K.; Zeng, P. Identification of circadian clock genes as regulators of immune infiltration in hepatocellular carcinoma. J. Cancer 2022, 13, 3199–3208. [Google Scholar] [CrossRef]
  84. Zhu, X.; Zhang, Z.; Zhang, J.; Xiao, Y.; Wang, H.; Wang, M.; Jiang, M.; Xu, Y. Single-cell and bulk transcriptomic analyses reveal a stemness and circadian rhythm disturbance-related signature predicting clinical outcome and immunotherapy response in hepatocellular carcinoma. Curr. Gene Ther. 2025, 25, 178–193. [Google Scholar]
  85. Zhang, Z.; Ma, F.; Zhou, F.; Chen, Y.; Wang, X.; Zhang, H.; Zhu, Y.; Bi, J.; Zhang, Y. Functional polymorphisms of circadian negative feedback regulation genes are associated with clinical outcome in HCC patients receiving radical resection. Med. Oncol. 2014, 31, 179. [Google Scholar] [CrossRef]
  86. Zhao, B.; Lu, J.; Yin, J.; Liu, H.; Guo, X.; Yang, Y.; Ge, N.; Zhu, Y.; Zhang, H.; Xing, J. A functional polymorphism in per3 gene is associated with prognosis in hepatocellular carcinoma. Liver Int. 2012, 32, 1451–1459. [Google Scholar]
  87. Yuan, P.; Wang, S.; Zhou, F.; Wan, S.; Yang, Y.; Huang, X.; Zhang, Z.; Zhu, Y.; Zhang, H.; Xing, J. Functional polymorphisms in the npas2 gene are associated with overall survival in transcatheter arterial chemoembolization-treated HCC patients. Cancer Sci. 2014, 105, 825–832. [Google Scholar] [CrossRef]
  88. Fey, R.M.; Billo, A.; Clister, T.; Doan, K.L.; Berry, E.G.; Tibbitts, D.C.; Kulkarni, R.P. Personalization of cancer treatment: Exploring the role of chronotherapy in immune checkpoint inhibitor efficacy. Cancers 2025, 17, 732. [Google Scholar] [CrossRef] [PubMed]
  89. Das, M.; Kumar, D.; Sauceda, C.; Oberg, A.; Ellies, L.G.; Zeng, L.; Jih, L.J.; Newton, I.G.; Webster, J.G. Time-restricted feeding attenuates metabolic dysfunction-associated steatohepatitis and hepatocellular carcinoma in obese male mice. Cancers 2024, 16, 1513. [Google Scholar] [PubMed]
  90. Becker, S.; Momoh, J.; Biancacci, I.; Mockel, D.; Wang, Q.; May, J.N.; Su, H.; Candels, L.S.; Berres, M.L.; Kiessling, F.; et al. Intermittent fasting primes the tumor microenvironment and improves nanomedicine delivery in hepatocellular carcinoma. Small 2023, 19, e2208042. [Google Scholar] [CrossRef]
  91. Molina-Aguilar, C.; Guerrero-Carrillo, M.J.; Espinosa-Aguirre, J.J.; Olguin-Reyes, S.; Castro-Belio, T.; Vazquez-Martinez, O.; Rivera-Zavala, J.B.; Diaz-Munoz, M. Time-caloric restriction inhibits the neoplastic transformation of cirrhotic liver in rats treated with diethylnitrosamine. Carcinogenesis 2017, 38, 847–858. [Google Scholar] [CrossRef]
  92. Meng, Q.J.; Logunova, L.; Maywood, E.S.; Gallego, M.; Lebiecki, J.; Brown, T.M.; Brown, T.M.; Sládek, M.; Semikhodskii, A.S.; Glossop, N.R.J. Setting clock speed in mammals: The ck1 epsilon tau mutation in mice accelerates circadian pacemakers by selectively destabilizing period proteins. Neuron 2008, 58, 78–88. [Google Scholar] [CrossRef]
  93. Ramsey, K.M.; Yoshino, J.; Brace, C.S.; Abrassart, D.; Kobayashi, Y.; Marcheva, B.; Hong, H.K.; Chong, J.L.; Buhr, E.D.; Lee, C.; et al. Circadian clock feedback cycle through nampt-mediated nad+ biosynthesis. Science 2009, 324, 651–654. [Google Scholar] [CrossRef]
  94. Ramanathan, C.; Kathale, N.D.; Liu, D.; Lee, C.; Freeman, D.A.; Hogenesch, J.B.; Cao, R.; Liu, A.C. mTOR signaling regulates central and peripheral circadian clock function. PLoS Genet. 2018, 14, e1007369. [Google Scholar]
  95. Krstic, J.; Reinisch, I.; Schindlmaier, K.; Galhuber, M.; Riahi, Z.; Berger, N.; Kupper, N.; Moyschwitz, E.; Auer, M.; Michenthaler, H.; et al. Fasting improves therapeutic response in hepatocellular carcinoma through p53-dependent metabolic synergism. Sci. Adv. 2022, 8, eabh2635. [Google Scholar] [CrossRef]
  96. Usman, M.; Javed, N.; Jawhari, A.; Ghouri, N.; Waqar, S.; Shah, F.; Ahmad, S.; Hart, A.; Hameed, B.; Khan, Q.M.; et al. Ramadan intermittent fasting for patients with gastrointestinal and hepatobiliary diseases: Practical guidance for health-care professionals. Lancet Gastroenterol. Hepatol. 2025, 10, 168–182. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, T.H.; Wu, C.H.; Yeh, C.T.; Su, S.C.; Hsia, S.M.; Liang, K.H.; Chen, C.C.; Hsueh, C.; Chen, C.Y. Melatonin suppresses hepatocellular carcinoma progression via lncrna-cps1-it-mediated hif-1alpha inactivation. Oncotarget 2017, 8, 82280–82293. [Google Scholar] [CrossRef] [PubMed]
  98. Nabih, H.K.; Hamed, A.R.; Yahya, S.M.M. Anti-proliferative effect of melatonin in human hepatoma hepg2 cells occurs mainly through cell cycle arrest and inflammation inhibition. Sci. Rep. 2023, 13, 4396. [Google Scholar] [CrossRef]
  99. Kim, J.I.; Cheon, H.G. Melatonin ameliorates hepatic fibrosis via the melatonin receptor 2-mediated upregulation of bmal1 and anti-oxidative enzymes. Eur. J. Pharmacol. 2024, 966, 176337. [Google Scholar] [CrossRef] [PubMed]
  100. Han, Y.; Chen, L.; Baiocchi, L.; Ceci, L.; Glaser, S.; Francis, H.; Alpini, G.; Kennedy, L. Circadian rhythm and melatonin in liver carcinogenesis: Updates on current findings. Crit. Rev. Oncog. 2021, 26, 69–85. [Google Scholar] [CrossRef]
  101. Carbajo-Pescador, S.; Ordonez, R.; Benet, M.; Jover, R.; Garcia-Palomo, A.; Mauriz, J.L.; Gonzalez-Gallego, J. Inhibition of vegf expression through blockade of hif1alpha and stat3 signalling mediates the anti-angiogenic effect of melatonin in hepg2 liver cancer cells. Br. J. Cancer 2013, 109, 83–91. [Google Scholar] [CrossRef]
  102. Prieto-Dominguez, N.; Mendez-Blanco, C.; Carbajo-Pescador, S.; Fondevila, F.; Garcia-Palomo, A.; Gonzalez-Gallego, J.; Mauriz, J.L. Melatonin enhances sorafenib actions in human hepatocarcinoma cells by inhibiting mtorc1/p70s6k/hif-1alpha and hypoxia-mediated mitophagy. Oncotarget 2017, 8, 91402–91414. [Google Scholar] [CrossRef]
  103. Carbajo-Pescador, S.; Garcia-Palomo, A.; Martin-Renedo, J.; Piva, M.; Gonzalez-Gallego, J.; Mauriz, J. Melatonin modulation of intracellular signaling pathways in HCC hepg2 cell line: Role of the mt1 receptor. J. Pineal Res. 2011, 51, 463–471. [Google Scholar] [CrossRef]
  104. Guo, R.; Rao, P.G.; Liao, B.Z.; Luo, X.; Yang, W.W.; Lei, X.H.; Ye, J.M. Melatonin suppresses pd-l1 expression and exerts antitumor activity in hepatocellular carcinoma. Sci. Rep. 2025, 15, 8451. [Google Scholar] [CrossRef]
  105. Ordonez, R.; Carbajo-Pescador, S.; Prieto-Dominguez, N.; Garcia-Palomo, A.; Gonzalez-Gallego, J.; Mauriz, J.L. Inhibition of matrix metalloproteinase-9 and nuclear factor kappa b contribute to melatonin prevention of motility and invasiveness in hepg2 liver cancer cells. J. Pineal Res. 2014, 56, 20–30. [Google Scholar] [CrossRef]
  106. Dauchy, R.T.; Wren-Dail, M.A.; Dupepe, L.M.; Hill, S.M.; Xiang, S.; Anbalagan, M.; Belancio, V.P.; Dauchy, E.M.; Blask, E. Effect of daytime blue-enriched led light on the nighttime circadian melatonin inhibition of hepatoma 7288ctc warburg effect and progression. Comp. Med. 2018, 68, 269–279. [Google Scholar] [CrossRef]
  107. Moreira, A.J.; Ordonez, R.; Cerski, C.T.; Picada, J.N.; Garcia-Palomo, A.; Marroni, N.P.; Mauriz, J.L.; González-Gallego, J. Melatonin activates ER stress and apoptosis in rats with diethylnitrosamine-induced hepatocarcinogenesis. PLoS ONE 2015, 10, e0144517. [Google Scholar] [CrossRef]
  108. Lin, S.; Hoffmann, K.; Gao, C.; Petrulionis, M.; Herr, I.; Schemmer, P. Melatonin promotes sorafenib-induced apoptosis through synergistic activation of jnk/c-jun pathway in human hepatocellular carcinoma. J. Pineal Res. 2017, 62, e12398. [Google Scholar] [CrossRef]
  109. Cheng, L.; Liu, J.; Liu, Q.; Liu, Y.; Fan, L.; Wang, F.; Yu, H.; Li, Y.; Bu, L.; Li, X.; et al. Exosomes from melatonin treated hepatocellularcarcinoma cells alter the immunosupression status through stat3 pathway in macrophages. Int. J. Biol. Sci. 2017, 13, 723–734. [Google Scholar] [CrossRef]
  110. Zou, Y.; Sun, H.; Guo, Y.; Shi, Y.; Jiang, Z.; Huang, J.; Li, L.; Jiang, F.; Lin, Z.; Wu, J.; et al. Integrative pan-cancer analysis reveals decreased melatonergic gene expression in carcinogenesis and rora as a prognostic marker for hepatocellular carcinoma. Front. Oncol. 2021, 11, 643983. [Google Scholar] [CrossRef]
  111. Sartorelli, L.S.; Neto, R.; Mosqueta-Pinheiro, M.; de Castro, T.B.; Fernandes, F.; Silva, R.; Novais, A.A.; Chuffa, L.G.D.A.; Reiter, R.J.; Zuccari, D.A.P.d.C. Blood melatonin level can serve as a potential biomarker for prostate and hepatocellular carcinomas. Melatonin Res. 2021, 4, 253–269. [Google Scholar] [CrossRef]
  112. Lorente, L.; Rodriguez, S.T.; Sanz, P.; Abreu-Gonzalez, P.; Gonzalez-Rivero, A.F.; Perez-Cejas, A.; Padilla, J.; Díaz, D.; González, A.; Martín, M.M.; et al. Low serum melatonin levels prior to liver transplantation in patients with HCC are associated with lower survival after liver transplantation. Int. J. Mol. Sci. 2019, 20, 1696. [Google Scholar]
  113. Levi, F.; Zidani, R.; Misset, J.L. Randomised multicentre trial of chronotherapy with oxaliplatin, fluorouracil, and folinic acid in metastatic colorectal cancer. International organization for cancer chronotherapy. Lancet 1997, 350, 681–686. [Google Scholar]
  114. Printezi, M.I.; Kilgallen, A.B.; Bond, M.J.G.; Stibler, U.; Putker, M.; Teske, A.J.; Cramer, M.J.; Punt, C.J.A.; Sluijer, J.P.G.; Huitema, A.D.R.; et al. Toxicity and efficacy of chronomodulated chemotherapy: A systematic review. Lancet Oncol. 2022, 23, e129–e143. [Google Scholar] [CrossRef]
  115. Voigt, R.M.; Forsyth, C.B.; Keshavarzian, A. Circadian rhythms: A regulator of gastrointestinal health and dysfunction. Expert. Rev. Gastroenterol. Hepatol. 2019, 13, 411–424. [Google Scholar] [PubMed]
  116. Yu, F.; Zhang, T.; Zhou, C.; Xu, H.; Guo, L.; Chen, M.; Wu, B. The circadian clock gene bmal1 controls intestinal exporter mrp2 and drug disposition. Theranostics 2019, 9, 2754–2767. [Google Scholar] [CrossRef] [PubMed]
  117. Iwasaki, M.; Koyanagi, S.; Suzuki, N.; Katamune, C.; Matsunaga, N.; Watanabe, N.; Takahashi, M.; Izumi, T.; Ohdo, S. Circadian modulation in the intestinal absorption of p-glycoprotein substrates in monkeys. Mol. Pharmacol. 2015, 88, 29–37. [Google Scholar] [CrossRef]
  118. Eckerbom, P.; Hansell, P.; Cox, E.; Buchanan, C.; Weis, J.; Palm, F.; Francis, S.; Liss, P. Circadian variation in renal blood flow and kidney function in healthy volunteers monitored with noninvasive MRI. Am. J. Physiol. Renal Physiol. 2020, 319, F966–F978. [Google Scholar] [CrossRef] [PubMed]
  119. Zhao, M.; Zhao, H.; Deng, J.; Guo, L.; Wu, B. Role of the clock protein in liver detoxification. Br. J. Pharmacol. 2019, 176, 4639–4652. [Google Scholar] [CrossRef]
  120. Lin, Y.; Wang, S.; Zhou, Z.; Guo, L.; Yu, F.; Wu, B. Bmal1 regulates circadian expression of cytochrome p450 3a11 and drug metabolism in mice. Commun. Biol. 2019, 2, 378. [Google Scholar] [CrossRef]
  121. Ge, W.; Wang, T.; Zhao, Y.; Yang, Y.; Sun, Q.; Yang, X.; Gao, Y.; Xu, X.; Zhang, J. Period1 mediates rhythmic metabolism of toxins by interacting with cyp2e1. Cell Death Dis. 2021, 12, 76. [Google Scholar] [CrossRef]
  122. Chen, M.; Chen, M.; Lu, D.; Wang, Y.; Zhang, L.; Wang, Z.; Wu, B. Period 2 regulates cyp2b10 expression and activity in mouse liver. Front. Pharmacol. 2021, 12, 764124. [Google Scholar] [CrossRef]
  123. Fekry, B.; Aggarwal, S.; Van Drunen, R.; Bravo, R.; Escalante, A.; Atkins, C.; Pan, S.; Chen, Z.; Sun, K.; Hall, D.R.; et al. Targeting the hepatic circadian clock concomitant with tyrosine kinase inhibition reverses late-stage hepatocellular carcinoma. Cell Rep. 2025, 44, 116313. [Google Scholar] [CrossRef]
  124. Hassan, S.A.; Ali, A.A.H.; Sohn, D.; Flogel, U.; Janicke, R.U.; Korf, H.W.; von Gall, C. Does timing matter in radiotherapy of hepatocellular carcinoma? An experimental study in mice. Cancer Med. 2021, 10, 7712–7725. [Google Scholar] [CrossRef]
  125. Yassine, M.; Hassan, S.A.; Sommer, S.; Yucel, L.A.; Bellert, H.; Hallenberger, J.; Sohn, D.; Korf, H.W.; von Gall, C.; Ali, A.A.H. Radiotherapy of the hepatocellular carcinoma in mice has a time-of-day-dependent impact on the mouse hippocampus. Cells 2022, 12, 61. [Google Scholar] [CrossRef] [PubMed]
  126. Karaboue, A.; Innominato, P.F.; Wreglesworth, N.I.; Duchemann, B.; Adam, R.; Levi, F.A. Why does circadian timing of administration matter for immune checkpoint inhibitors’ efficacy? Br. J. Cancer 2024, 131, 783–796. [Google Scholar] [CrossRef]
  127. Lim, M.; Espinoza, M.; Huang, Y.H.; Franses, J.; Zhu, H.; Hsiehchen, D. Complete response to immunotherapy in patients with hepatocellular carcinoma. JAMA Netw. Open 2025, 8, e2461735. [Google Scholar] [CrossRef]
  128. Shek, D.; Read, S.A.; Nagrial, A.; Carlino, M.S.; Gao, B.; George, J.; Ahlenstiel, G. Immune-checkpoint inhibitors for advanced hepatocellular carcinoma: A synopsis of response rates. Oncologist 2021, 26, e1216–e1225. [Google Scholar] [CrossRef]
  129. Landre, T.; Karaboue, A.; Buchwald, Z.S.; Innominato, P.F.; Qian, D.C.; Assie, J.B.; Chouaid, C.; Levi, F.; Duchemann, B. Time-dependent efficacy of immune checkpoint inhibitors in the treatment of metastatic cancers: A meta-analysis. J. Clin. Oncol. 2024, 41, 2562. [Google Scholar] [CrossRef]
  130. Kemeny, M.M.; Alava, G.; Oliver, J.M. Improving responses in hepatomas with circadian-patterned hepatic artery infusions of recombinant interleukin-2. J. Immunother. 1992, 12, 219–223. [Google Scholar] [CrossRef] [PubMed]
  131. Naganuma, A.; Kakizaki, S.; Hatanaka, T.; Hiraoka, A.; Tada, T.; Hirooka, M.; Karyiama, K.; Tani, J.; Atsukawa, M.; Takaguchi, K.; et al. Impact of time-of-day atezolizumab plus bevacizumab combination therapy infusion for unresectable HCC: A retrospective multicenter study. Hepatol. Res. 2025, 55, 741–751. [Google Scholar] [CrossRef] [PubMed]
  132. Centanni, M.; Moes, D.; Troconiz, I.F.; Ciccolini, J.; van Hasselt, J.G.C. Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors. Clin. Pharmacokinet. 2019, 58, 835–857. [Google Scholar] [CrossRef] [PubMed]
  133. Qian, D.C.; Buchwald, Z.S. Immune checkpoint inhibitors and timing of administration—authors’ reply. Lancet Oncol. 2022, 23, e57. [Google Scholar] [CrossRef]
  134. van der Veen, E.L.; Giesen, D.; Pot-de Jong, L.; Jorritsma-Smit, A.; De Vries, E.G.E.; Lub-de Hooge, M.N. 89zr-pembrolizumab biodistribution is influenced by pd-1-mediated uptake in lymphoid organs. J. Immunother. Cancer 2020, 8, e000938. [Google Scholar] [CrossRef]
  135. Wang, C.; Zeng, Q.; Gul, Z.M.; Wang, S.; Pick, R.; Cheng, P.; Bill, R.; Wu, Y.; Naulaerts, S.; Barnoud, C.; et al. Circadian tumor infiltration and function of cd8(+) t cells dictate immunotherapy efficacy. Cell 2024, 187, 2690–2702.e17. [Google Scholar] [CrossRef]
  136. He, W.; Holtkamp, S.; Hergenhan, S.M.; Kraus, K.; de Juan, A.; Weber, J.; Bradfield, P.; Grenier, J.M.P.; Pelletier, J.; Druzd, D.; et al. Circadian expression of migratory factors establishes lineage-specific signatures that guide the homing of leukocyte subsets to tissues. Immunity 2018, 49, 1175–1190.e7. [Google Scholar] [CrossRef]
  137. Gibbs, J.; Ince, L.; Matthews, L.; Mei, J.; Bell, T.; Yang, N.; Saer, B.; Begley, N.; Poolman, T.; Pariollaud, M.; et al. An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat. Med. 2014, 20, 919–926. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular pathways of circadian clocks. The heterodimer BMAL1-CLOCK is a transcription factor for circadian-controlled genes including PER, CRY, DEC, REV-ERB, and ROR. Two autoregulatory feedback loops inhibits BMAL1-CLOCK action, either by direct inhibition (the heterodimer PER-CLOCK) or by competition with DNA binding (DEC). A third loop modulates BMAL1 expression, either promoting (ROR) or repressing it (REV-ERB). BMAL-1, brain and muscle ARNT-like-1; CLOCK, circadian locomotor output cycle kaput; PER, period; CYR, cryptochrome; DEC, differentiation embryo chondrocytes; RORs, retinoic acid-related orphan nuclear receptors. E-box and RORE cis regulatory elements. Green arrows indicate promotion of transcription and red ones inhibition.
Figure 1. Molecular pathways of circadian clocks. The heterodimer BMAL1-CLOCK is a transcription factor for circadian-controlled genes including PER, CRY, DEC, REV-ERB, and ROR. Two autoregulatory feedback loops inhibits BMAL1-CLOCK action, either by direct inhibition (the heterodimer PER-CLOCK) or by competition with DNA binding (DEC). A third loop modulates BMAL1 expression, either promoting (ROR) or repressing it (REV-ERB). BMAL-1, brain and muscle ARNT-like-1; CLOCK, circadian locomotor output cycle kaput; PER, period; CYR, cryptochrome; DEC, differentiation embryo chondrocytes; RORs, retinoic acid-related orphan nuclear receptors. E-box and RORE cis regulatory elements. Green arrows indicate promotion of transcription and red ones inhibition.
Curroncol 33 00032 g001
Figure 2. Molecular pathways interlinked between circadian rhythms and hepatocarcinogenesis.
Figure 2. Molecular pathways interlinked between circadian rhythms and hepatocarcinogenesis.
Curroncol 33 00032 g002
Table 1. Epidemiological studies on the relationship between circadian deregulation and HCC.
Table 1. Epidemiological studies on the relationship between circadian deregulation and HCC.
StudyStudy DesignFactors AdjustedMain Results
Geographical longitude and HCC incidence
Gu, F.
2017 [50]
SEER data (2000–2012) from 607 counties across 11 U.S. states.
Incidence rates for 23 cancers types.
AgePer 5° westward increase in longitude within a time zone: HCC RR 1.11
[95%CI 1.05–1.18]
VoPham, T.
2018 [49]
SEER data (2000–2014) from 16 population-based US cancer registries.
56,347 HCC cases included.
Demographics
Socioeconomic status
Obesity, T2DM rates
Lifestyle habits
Shift work occupation
Environmental factors
Per 5° westward increase in longitude within a time zone: HCC IRR 1.07
[95%CI 1.01–1.14]
Niu, J.
2024 [51]
State Cancer Profiles (2016–2020) from 2853 counties across 48 U.S. states.
Incidence rates for 8 cancers types.
Demographics
Socioeconomic status
Obesity rate
Altitude
Environmental factors
Decreasing incidence of liver and bile duct cancers from west to east within a time zone
Residential outdoor LAN exposure (measured from satellite imagery) and HCC risk
Park, Y.
2022 [53]
U.S. prospective cohort of 451,945 participants aged 50–71 years from the NIH-AARP Diet and Health Study (1995–1996).
Median follow up 12.2 years.
Demographics
Socioeconomic status
BMI
Lifestyle habits
No association between outdoor LAN exposure and HCC risk: RR 0.96 [95%CI 0.77–1.20], quartile 5 vs. 1
Night shift work and HCC risk
Arafa, A.
2021 [55]
Japanese prospective cohort of 45,390 workers aged 40–79 years from the JACC Study (1988–1990).
Median follow up 14.2 years.
Demographics
Socioeconomic status
Lifestyle habits
Rotation shift-work associates with increased HCC risk:
HR 0.54 [95%CI 0.30–0.98]
Sleep patterns and HCC risk
Hu, L.Y.
2013 [8]
Taiwan prospective cohort of 63,381 newly diagnosed sleep disorder patients from the Taiwan National Health Insurance Research Database (1996–2010).
Median follow up 6.2 years.
DemographicsSIR for liver cancer 1.44
[95%CI 1.28–1.61]
Royse, K.E.
2017 [59]
U.S. prospective cohort of 139,368 postmenopausal women from the
Prospective Women’s Health Institute Study (1993–1998). Median follow up 13.8 years.
Demographics
Socioeconomic status
Lifestyle habits
BMI and T2DM
HCC HR compared with 6–8 H sleep:
≤5 H: 1.10 [95%CI 0.63–1.94]
6 H: 0.90 [95%CI 0.61–1.32]
≥9 H: 1.88 [95%CI 1.02–3.45]
Long, L.
2023 [58]
U.S. prospective cohort of 295,837 participants from the NIH-AARP Diet and Health Study (1995–1996).
Median follow up 15.5 years.
Demographics
Socioeconomic status
Lifestyle habits
HCC HR compared with 7–8 H sleep:
<5 H: 2.00 [95%CI 1.22–3.26]
5–6 H: 1.15 [95%CI 0.91–1.45]
≥9 H: 1.63 [95%CI 1.04–2.65]
Napping > 1 H: 1.46 [95%CI 1.04–2.06]
Wang, W.
2024 [60]
UK prospective cohort of 408,560 participants free of CLD, aged 40–70 years from the UK Biobank (2006–2010).
Median follow up 12.5 years.
Demographics
Lifestyle habits
BMI, lipid profile, HTN and T2DM
Healthy sleep score associates with decreased HCC risk: HR 0.53
[95%CI 0.37–0.77]
Wang, Q.
2024 [57]
UK prospective cohort of 489,261 participants aged 37–73 years from the UK Biobank (2006–2010).
Median follow up 13.8 years.
Demographics
Socioeconomic status
BMI, T2DM, HTN
Lifestyle habits
HCC HR compared with 7 H sleep:
≤5 H: 1.26 [95%CI 0.98–1.60]
6 H: 1.02 [95%CI 0.86–1.24]
8 H: 1.04 [95%CI 0.89–1.20]
≥9 H: 1.23 [95%CI 1.01–1.51]
BMI, body mass index; CI, confidence interval; CLD, chronic liver disease; HCC, hepatocellular carcinoma; HR, hazard ratio; HTN, hypertension; IRR, incidence rate ratio; JACC, Japanese Collaborative Cohort; LAN, light-at-night; NIH-AARP, National Institute of Health—American Association of Retired Persons; RR, relative risk; SEER, Surveillance, Epidemiology, and End Results; SIR, standardized incidence ratio; T2DM, type 2 diabetes mellitus.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Machado, M.V. Hepatocellular Carcinoma Around the Clock. Curr. Oncol. 2026, 33, 32. https://doi.org/10.3390/curroncol33010032

AMA Style

Machado MV. Hepatocellular Carcinoma Around the Clock. Current Oncology. 2026; 33(1):32. https://doi.org/10.3390/curroncol33010032

Chicago/Turabian Style

Machado, Mariana Verdelho. 2026. "Hepatocellular Carcinoma Around the Clock" Current Oncology 33, no. 1: 32. https://doi.org/10.3390/curroncol33010032

APA Style

Machado, M. V. (2026). Hepatocellular Carcinoma Around the Clock. Current Oncology, 33(1), 32. https://doi.org/10.3390/curroncol33010032

Article Metrics

Back to TopTop