Next Article in Journal
CT Fluoroscopy-Guided Percutaneous Gastrostomy in the Palliative Management of Advanced and Relapsed Ovarian Cancer: The Charité Experiences and a Review of the Literature
Next Article in Special Issue
Impact of Hypoxia on Radiation-Based Therapies for Liver Cancer
Previous Article in Journal
Selinexor in Combination with Decitabine Attenuates Ovarian Cancer in Mice
Previous Article in Special Issue
Functional Imaging of Hypoxia: PET and MRI
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Antitumor Activity of Metformin Combined with Locoregional Therapy for Liver Cancer: Evidence and Future Directions

1
School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
2
Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
3
Department of Radiology, Division of Interventional Radiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(18), 4538; https://doi.org/10.3390/cancers15184538
Submission received: 17 August 2023 / Revised: 6 September 2023 / Accepted: 7 September 2023 / Published: 13 September 2023
(This article belongs to the Special Issue Liver Cancers Molecular Biomarkers Predicting Outcome)

Abstract

:

Simple Summary

Existing therapies for hepatocellular carcinoma, although effective, often have limitations in their long-term efficacy. Metformin is increasingly being recognized for its anti-cancer properties and potential in augmenting the tumor response following locoregional therapy. This review analyzed existing comparative studies on combination therapy with metformin and found that metformin had the potential to augment the effects of current locoregional therapies for HCC management.

Abstract

This article aimed to examine the effect of metformin use on improving outcomes after liver-directed therapy in patients with HCC and identify future directions with the adjuvant use of and potential therapeutic agents that operate on similar mechanistic pathways. Databases were queried to identify pertinent articles on metformin’s use as an anti-cancer agent in HCC. Eleven studies were included, with five pre-clinical and six clinical studies. The mean overall survival (OS) and progression-free survival were both higher in the locoregional therapy (LRT) + metformin-treated groups. The outcome variables, including local tumor recurrence rate, reduction in HCC tumor growth and size, tumor growth, proliferation, migration and invasion of HCC cells, HCC cell apoptosis, DNA damage, and cell cycle arrest, showed favorable outcomes in the LRT + metformin-treated groups compared with LRT alone. This systemic review provides a strong signal that metformin use can improve the tumor response after locoregional therapy. Well-controlled prospective trials will be needed to elucidate the potential antitumor effects of metformin and other mTOR inhibitors.

1. Introduction

Since 1980, the incidence of liver cancer has more than tripled and death rates have doubled, indicating a clear need for aggressive diagnostic and treatment modalities to reduce the burden of this disease in the United States and worldwide. Hepatocellular carcinoma (HCC) is classified as the most common type of liver cancer, accounting for approximately 80% of all diagnoses [1]. It also is a leading cause of cancer mortality worldwide, with a 5-year survival of under 20% [2,3,4]. Although studies show a plateau or even a decrease in the number of new HCC cases in recent years [2,3], previous Surveillance, Epidemiology, and End Results (SEER) database analyses have projected that there will still be over 56,000 cases of HCC by 2030 in the US and 22 million cases in the next two decades worldwide [4,5]. The most common risk factors associated with the development of this malignancy include viral hepatitis, primarily Hepatitis B and C, alcoholic liver disease and cirrhosis, and, more recently, non-alcoholic fatty liver disease [6,7]. Several studies have also identified a correlation between metabolic syndrome, male gender, and Hispanic and African Americans with higher incidences of hepatocellular carcinoma diagnosis [8].
The plateau in hepatocellular carcinoma cases in recent years is likely attributed to more aggressive viral hepatitis prevention measures, rather than successful and efficient treatment interventions for HCC [9], which highlights that, although there may be implemented strategies to reduce the number of new HCC cases, the staggering mortality rate shows that there is still limited treatment success in patients who are suffering the consequences of HCC [10]. The 2-year survival rates in the United States have remained under 50%, and 5-year survival rates of HCC remain below 12% [11,12].
There are various treatment options for HCC, including liver transplantation, curative resection, radiofrequency ablation (RFA), chemoembolization, and systemic targeted agents. The treatment of the cancer depends on the stage of the tumor at diagnosis, patient performance status, and liver function, requiring a holistic evaluation of the patient before deciding on the best treatment modality. Surgical resection and liver transplantation offer the best curative treatments for HCC [13]; however, eligibility for these procedures requires detection at an early stage, and, unfortunately, the majority of patients tend to be diagnosed at advanced disease stages [2,14,15,16,17,18,19,20]. In patients with more advanced disease, other treatments, such as radiofrequency ablation (RFA), transarterial chemoembolization (TACE), and yttrium 90 (Y90) radioembolization, have shown to be successful at reducing tumor progression, with radioembolization having the safest toxicity profile [20,21,22,23,24]. Some studies have shown that these treatments are equally effective in achieving tumor necrosis [25,26,27], while others identified that Y90 radioembolization leads to significantly higher survival rates in comparison with transarterial chemoembolization [28,29]. Despite recent advances in systemic and locoregional therapies, prognosis for many patients with unresectable HCC remains poor due to limited responses and a high rate of recurrence. The global disease burden and poor outcomes associated with HCC have prompted efforts to identify strategies to improve responses to existing therapies, including the use of adjunctive agents with liver-directed therapies (LDTs) [30,31,32,33,34,35].
Researchers have reported improved outcomes associated with the use of various adjuvant drugs combined with locoregional therapies, such as longer overall survival and delayed HCC tumor recurrence when LRT was combined with angiotensin II receptor 1 blockers (sartans) [36] and angiotensin-converting enzyme inhibitors (ACE-I) [37,38,39]. Additionally, adjuvant use of antiviral agents with LRT has been of interest for decades, as viral hepatitis is a primary driver of a significant proportion of patients with HCC, and the use of such drugs has been associated with improved overall and recurrence-free survival [40,41,42,43,44,45,46,47,48]. Vitamin K analogs and retinoids have also shown promise as an adjuvant therapy against HCC, given various anti-tumor effects, including the induction of cell cycle arrest and inhibition of HCC cell proliferation [38,49,50,51,52,53,54,55].
The interest in understanding the role of metformin in cancer prevention was first sparked by an observational study conducted in 2005 that showed its ability to suppress the incidence and progression of various cancers through several mechanisms, including the inhibition of insulin-like growth factor-1 (IGF-1) [56], activation of the adenosine 5′-monophosphate-activated protein kinase (AMPK) [57], inhibition of the mammalian target of rapamycin complex I (mTORC1) [58], inhibition of nuclear factor-kB (NF-kB) [59], and inhibition of oxidation phosphorylation in cancer cells [60,61]. Subsequent studies have shown a protective effect of metformin in preventing the onset of liver cancer, specifically hepatocellular carcinoma [62,63,64,65,66,67,68,69,70]; however, further investigation is warranted to identify the true capacity of metformin in the setting of hepatocellular carcinoma. A growing number of population studies and meta-analyses have found that metformin use is associated with decreased incidence and mortality for various malignancies, including prostate, endometrial, gastric, pancreatic, thyroid, breast cancer, and colon cancer [71,72,73,74,75]. Recent preclinical and clinical studies indicate that metformin use plays a chemo-preventative role in HCC development through mTOR inhibition [69,76,77,78]. Additionally, the utilization of stereotactic body radiation therapy (SBRT) has shown promising results in cases of inoperable HCC [79,80], highlighting the potential of external-beam radiotherapies as a novel locoregional therapy that can also be combined with adjunctive agents.
Metformin use shows particular promise in HCC, with large studies across various geographic locations and clinical settings showing that its use is associated with reduced incidence of HCC in patients with cirrhosis, as well as improved prognosis among patients with HCC. This systematic review compiles current literature regarding the effect of metformin use on improving outcomes after liver-directed therapy in patients with HCC. Future directions regarding the adjuvant use of metformin and potential therapeutic agents that work on similar mechanistic pathways will be discussed.

2. Materials and Methods

A systematic review was performed on the existing literature examining metformin as an adjuvant agent in patients with hepatocellular carcinoma undergoing locoregional therapy. All of the literature search findings were reported in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. Since no human subjects were studied in this research, this systematic review was exempt from official Institutional Review Board (IRB) approval. All studies were uploaded to EndNote and were screened using the Covidence systematic review software (Veritas Health Innovation, Melbourne, Australia, www.covidence.org. URL accessed on 27 December 2022). This review was not registered in PROSPERO.

2.1. Literature Search and Strategy

The PubMed, Cochrane, EMBASE, and MEDLINE library databases were each queried in December 2022. Each database was searched for any relevant studies pertaining to metformin’s use as an anti-cancer agent in HCC. Articles published between 1981 and 2022 were analyzed. The search terms for this systematic review contained the following combinations of terms: “metformin”, “hepatocellular carcinoma”, “drug response”, “type 2 diabetes mellitus”, “tumor sensitizing”, “locoregional therapy”, “transarterial chemoembolization”, “transarterial radioembolization”, “radiofrequency thermal ablation”, “yttrium-90 radioembolization”, and “external beam radiation therapy”. The reference sections of each eligible full-text article were reviewed to ensure that additional articles were not missed during the initial database literature search.

2.2. Eligibility and Search Criteria

Articles were selected by first reviewing the abstracts for relevance, followed by full-text assessment according to the stated inclusion and exclusion criteria. All duplicate articles were removed in Covidence during the initial screening process.
The eligibility criteria for this systematic review included the following: (1) the study enrolled adult patients (>18 years); (2) the patients in the study had a diagnosis of hepatocellular carcinoma; (3) the study consisted of patients who underwent locoregional therapy for HCC, including radiofrequency thermal ablation (RFA), yttrium-90 (Y90) radioembolization, transarterial chemoembolization (TACE), transarterial radioembolization (TARE), microwave ablation (MWA), and external-beam radiotherapy (EBRT), including conventional external beam radiation, stereotactic body radiotherapy (SBRT), hypofractioned radiotherapy (HypoRT), and particle therapy (gamma ray or neutron ray); (4) patients were concomitantly taking metformin during LDT. Any studies that were (1) case series including fewer than four patients, (2) case reports including fewer than four patients, (3) reviews, letters, editorials, or animal studies, or (4) did not report relevant outcomes were excluded during the initial screening of titles and abstracts. Studies that reported outcomes on patients undergoing liver resection, transplantation, or systemic chemotherapies were also excluded.

2.3. Data Extraction

Data extraction was performed on all articles that were deemed eligible following full-text assessment. A custom table was generated in Microsoft Excel (version 2019; Microsoft, Redmond, VA, USA) to organize all contents of the data extraction. The following information was collected through data extraction: title, author, publication year, location of the study performed, number of patients, male-to-female ratio, age of patients, control and experimental groups, human cell type (if applicable), metformin dosage, study outcomes, overall survival, and progression-free survival.

2.4. Statistical Analysis

Aggregate data of all outcomes were collected, and the average values were calculated using the weighted average mean approach. If there was an instance where only the median and interquartile range of an outcome variable was provided, the average was calculated using Hozo’s formula [81]. All standard deviation calculations were performed using Hozo’s pooled standard deviation formula [81].

3. Results

3.1. Study Selection

There were 340 studies initially identified through the database search. Following title and abstract screening, there were 154 studies deemed eligible for full-text assessment. Eleven studies were ultimately included in the systematic review [82,83,84,85,86,87,88,89,90,91]. One study included both pre-clinical and clinical data and was thus considered two separate studies [89]. Figure 1 shows a PRISMA diagram outlining the selection process for eligible studies and reasons for exclusion.

3.2. Study and Patient Characteristics

Both pre-clinical and clinical studies were identified through the literature search process. Of the 11 total studies included in this systematic review, 5 studies were classified as human preclinical studies [87,88,89,90,91] and 6 studies were classified as human clinical studies [82,83,84,85,86,89]. All included studies were full-text publications and were published between 2012 and 2022. Among the 11 studies, 2 studies performed radiofrequency ablation (RFA) [82,90], 3 studies performed transarterial chemoembolization (TACE) [83,86,89], and 1 study performed Y90 radioembolization (Y90 RS) [84]. Four of the eleven studies performed external beam radiation therapies, which included stereotactic body radiotherapy (SBRT)/hypofractionated radiotherapy (HypoRT) (one study) [85], gamma-ray radiation/neutron radiation (two studies) [87,91], and ionizing radiation (one study) [88]. There was a total of 805 patients among the six clinical studies, with males representing 50.6% of total patients. A total of 581 patients were in the control group (locoregional therapy only) in the studies and 233 patients were in the experimental group (locoregional therapy + metformin combination therapy) in the studies. The mean age of patients was 65.6 ± 9.7 years (Table 1).

3.3. Effect of Metformin and Locoregional Therapy on Survival in Patients

There were four clinical studies reporting outcomes related to overall survival [82,83,85,89] and four clinical studies reporting outcomes on progression-free survival [82,83,85,86] (Table 2). Overall survival ranged from 12 months to 75 months, with a mean overall survival of 39.8 months in the locoregional therapy-treated group alone, and overall survival ranged from 12 months to 79 months with a mean overall survival of 39.1 months in the locoregional therapy + metformin-treated group. (Table 2). The progression-free survival ranged from 7 months to 74 months, with a mean progression-free survival of 35.5 months in the locoregional therapy-treated group alone, and progression-free survival ranged from 12 months to 79 months with a mean progression-free survival of 37.2 months in the locoregional therapy + metformin-treated group. (Table 2). Metformin + locoregional therapy resulted in improved overall survival (p < 0.001) and progression-free survival (p < 0.001) compared with locoregional therapy alone.

3.4. Tumor Recurrence

Two clinical studies analyzed tumor recurrence after either TACE alone or TACE + metformin combination therapy [83,86]. Both clinical studies reported lower local tumor recurrence in TACE + metformin-treated patients compared with TACE monotherapy (Table 2). Jung et al. 2022 reported local tumor recurrence of 85.2% in non-metformin-treated patients and 74.8% in metformin-treated groups. Chen 2022 reported that tumor recurrence for non-metformin- and metformin-treated groups was 52.39% vs. 29.19% at 1 year, 17.03% vs. 0.00% at 2 years, and 4.26% vs. 0.00% at 3 years.

3.5. Tumor Growth and Size

One clinical study (Table 2) [84] and two pre-clinical studies (Table 3) [34,90] reported outcomes related to tumor size and growth. All studies reported that combination therapy resulted in a greater reduction in HCC tumor growth and size in comparison with monotherapy. Similarly, Elsayed et al. 2021 reported changes in tumor growth as a change in total tumor diameter (TTD), revealing metformin + TACE was associated with a greater mean TTD reduction of 40.4 mm, compared with 17.1 mm in patients receiving TACE alone. (p = 0.018).

3.6. Tumor Proliferation, Migration, and Invasion

Two pre-clinical studies reported outcomes related to HCC cell proliferation, migration, and invasion (Table 3) [89,90]. Cell proliferation is defined as the rate of local tumor growth, migration is the directed movement of cells in response to chemotactic signals, and invasion is the vascular invasion of cancerous cells. All studies stated that a combination regimen of metformin use was associated with decreased tumor growth, proliferation, migration, and invasion of HCC cells in comparison with LRT alone.

3.7. Tumor Apoptosis, DNA Damage, and Cell Cycle Arrest

Two pre-clinical studies reported outcomes related to HCC cell apoptosis, DNA damage, and cell cycle arrest (Table 3) [89,93]. Both reported increased apoptosis, DNA damage, and cell cycle arrest in metformin combination therapy compared with LRT alone.

4. Discussion

This systematic review includes both preclinical and clinical data suggesting that metformin use may improve outcomes when used in conjunction with locoregional therapies. The preclinical studies analyzed in this review collectively found that metformin use could sensitize tumors to locoregional therapy, specifically by impairing several tumorigenic properties, including cell proliferation and migration. In addition, this systematic review is the first known to include human clinical data on locoregional therapy + metformin, compared with prior review articles that only included studies in animal models [94].
The results of our study found multiple preclinical and clinical outcomes supporting the utilization of metformin to augment the tumor response in patients with hepatocellular carcinoma. The six included clinical studies demonstrated superior survival outcomes in metformin plus locoregional therapy compared with locoregional monotherapy alone. We found that both the mean overall survival and mean progression-free survival were statistically higher in combination therapy-treated patients. When analyzing the effects of combination therapy on tumor recurrence, statistical analysis confirmed that the combination therapy with TACE and metformin resulted in statistically lower tumor recurrence rates compared with TACE monotherapy alone. Similarly, the comparison of tumor recurrence at 1 year, 2 years, and 3 years showed lower tumor recurrence percentages in metformin + locoregional therapy combination groups compared with locoregional therapy alone. Additional studies with various locoregional therapy may further support these results, as there were no identified studies that analyzed additional LRT + metformin specifically on tumor recurrence rates. All studies that compared variables correlating with tumor size and growth showed that combination therapy with metformin and locoregional therapy resulted in less tumor growth and smaller tumor sizes compared with patients treated with locoregional therapy alone. Preclinical studies reported data on the remaining variables, including tumor proliferation, migration, invasion, apoptosis, DNA damage, and cell cycle arrest. All preclinical studies reporting data on these variables identified superior outcomes with combination therapy in comparison with monotherapy alone. The combination of these results shows superior outcomes with metformin combination therapy when studying a variety of markers correlating to tumor growth progression.
Interestingly, there are several published literature reviews on the synergistic effects of metformin in treating other types of cancers. One published review found that, in patients with early stage colorectal cancer, metformin use was associated with significant benefits in recurrence-free survival, overall survival, and cancer-specific survival [95]. Additionally, this review found that, in patients with early stage prostate cancer, metformin was also associated with significant benefits in recurrence-free survival, overall survival, and cancer-specific survival [95]. This review, however, found that there were no significant benefits of metformin in treatment for breast and urothelial cancer. Another meta-analysis specifically analyzing the role of metformin in colorectal cancer management showed that patients already taking metformin had a significantly lower incidence of colorectal cancer. This review also determined that overall survival and cancer-specific survival were significantly higher in patients taking metformin compared with those who were not [96]. One study interestingly performed a study to estimate the overall cancer risk of type 2 diabetic patients already taking metformin. The study showed that patients taking metformin had a significantly lower risk of developing cancer, highlighting that metformin may be an independent protective factor for cancer risk entirely [97]. Another systematic review combined papers that, overall, supported the notion that metformin has clinical benefits in the treatment of gynecologic cancers, specifically cervical cancer [98]. Despite the results of prior systematic reviews, there is emerging literature on combination therapy with PD-1 inhibition plus rapamycin and metformin, which has been shown to enhance the anti-tumor efficacy in triple-negative breast cancer, highlighting promising therapies in years to come [99]. The ability of metformin to augment therapies in several cancer types highlights the versatility of the drug, as well as the legitimacy of its therapeutic benefits.
Although not fully understood, it is hypothesized that the major driving mechanism of metformin’s anti-cancer activity is likely related to the inhibition of the mammalian target of rapamycin (mTOR) pathway. There are two complexes within the mTOR pathway, mTORC1 and mTORC2, which both contain a catalytic subunit known as mTOR. The two complexes are known to receive stimuli from various hormonal signals throughout the body and are known to be imperative for cellular growth processes. mTORC1 primarily receives signals from pathways, including insulin-like growth factor 1 (IGF-1), insulin-like growth factor 2 (IGF-2), and AMP-activated protein kinase (AMPK). The mTOR pathway plays a key role in nutrient-sensing and regulation of various cellular processes, including cell proliferation, DNA repair, and protein synthesis. Metformin is found to specifically inhibit mTORC1 via the AMPK pathway through the activation of the tumor suppressor genes TSC1 and TSC2, which code for the tuberin and hamartin proteins, respectively. Additionally, metformin inhibits mTORC1 directly via AMPK with the aid of AMPK-inhibiting RAPTOR (regulatory-associated protein of mTOR) [100]. AMPK phosphorylates RAPTOR, and this phosphorylation is required for the inhibition of the mTORC1 complex. RAPTOR is an adaptor protein and positive regulator within the mTORC1 complex. The overactivation of mTOR is also associated with the development of various cancers, thus creating an opportunity to target this pathway with metformin and other therapeutic agents [101,102]. Metformin has also been shown to induce p53, which is a tumor-suppressor protein with the ability to also inhibit mTORC1. p53 is able to detect DNA damage and other genotoxic stresses that change the genetic material of cells. As a result, p53 is stimulated so that cellular growth and proliferation are halted to prevent the progression of abnormal cell growth. p53 also activates AMPK, and subsequently TSC1 and TSC2, which then inhibit mTORC1 [103]. Finally, metformin has also been found to reduce the production of fatty acid synthase, which is known to be oncogenic and significantly upregulated in cancer cells. Metformin studies have also found that it is able to reduce the production of reactive oxygen species, thus reducing oxidative stress and DNA damage through the inhibition of mitochondrial complexes [104].
The major role of mTOR in tumorigenesis has prompted the further investigation of potentially more potent mTOR inhibitors. Emerging literature suggests that the mTOR inhibitors anthramycin and Torin-2, for example, may be effective in treating HCC and suppressing liver cancer stem cells [45,105]. The efficacy of these agents may be further improved by loading them in specialized delivery systems, such as nanoparticle beads. The development of nanoparticles to deliver metformin to cancer cells has become of increasing interest, as the particles have the potential to successfully deliver the drug directly to the cancer cell of interest without systemic absorption or adverse side effects. A study conducted by Snima et al. analyzed the ability of O-carboxymethyl chitosan nanoparticles for the delivery of metformin to pancreatic cancer cells [106]. The study reported that cytotoxicity studies showed that the drug-incorporated nanoparticles induced statistically significant toxicity in the pancreatic cancer cells compared with normal healthy cell lines. Additionally, metformin-loaded nano-particles successfully demonstrated non-specific internalization by both normal and pancreatic cancer cells; however, the metformin released by nanoparticles preferentially induced toxicity in the pancreatic cancer cells, rather than the healthy cells.
This systematic review had several inherent limitations, primarily related to the methodology of the included studies. The included studies had limited data regarding the stage, extent of disease burden, and tumor biology, which precludes the ability to perform certain sub-analyses and further elucidate the effect of metformin. Similarly, there is likely significant heterogeneity regarding patient selection and how the locoregional therapy was technically performed, so the lack of standardization may have influenced the analysis in this study. Another limitation is that older studies may not have utilized contemporary treatment approaches, such as modern dosing strategies using Y90 radioembolization, which may have resulted in worse outcomes and obscured the full therapeutic effect of metformin. This systematic review is also limited by the fact that metformin is typically prescribed to patients with diabetes mellitus, which may introduce multiple layers of confounding bias. Additional studies on patients who do not have a concomitant diagnosis of diabetes mellitus may help to further understand the true efficacy of metformin as an adjuvant agent. Finally, the usage of metformin is rarely associated with life-threatening hepatotoxicity; therefore, patients being treated with metformin in the setting of hepatocellular carcinoma should be closely monitored for signs of acute liver failure. The included studies did not report any cases of life-threatening hepatotoxicity associated with metformin use; however, prospective trials would be useful to elucidate potential hepatotoxicity in this study.
Finally, this systemic review does not address the potential interactions between metformin and the recently approved checkpoint inhibitors. Emerging evidence suggests that metformin may augment the effect of checkpoint inhibitors [107]. Therefore, studies investigating the combination of these agents with locoregional therapy warrant further investigation.

5. Conclusions

Despite the limitations of this study, this systemic review provides a strong indication that metformin use can improve the tumor response after locoregional therapy. We found robust outcomes following combination therapy with metformin and locoregional therapy, including improved overall survival, reduced tumor recurrence, decreased cell proliferation and migration, and increased cancer cell DNA damage and apoptosis. Well-controlled prospective trials will be needed to elucidate the full potential antitumor effects of metformin and other mTOR inhibitors. Additional studies may help to fully understand the survival advantage suggested by this study.

Author Contributions

Conceptualization, M.E. and N.K.; Methodology, E.J.C.; Validation, E.J.C., M.E. and N.K.; Formal Analysis, E.J.C.; Investigation, E.J.C. and M.E.; Resources, E.J.C.; Data Curation, E.J.C. Writing—Original Draft Preparation, E.J.C.; Writing—Review and Editing, E.J.C., M.E. and N.K., Visualization, E.J.C., M.E. and N.K.; Supervision, M.E. and N.K.; Project Administration, N.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef]
  3. Rich, N.E.; Yopp, A.C.; Singal, A.G.; Murphy, C.C. Hepatocellular Carcinoma Incidence Is Decreasing Among Younger Adults in the United States. Clin. Gastroenterol. Hepatol. 2020, 18, 242–248.e245. [Google Scholar] [CrossRef]
  4. Petrick, J.L.; Kelly, S.P.; Altekruse, S.F.; McGlynn, K.A.; Rosenberg, P.S. Future of Hepatocellular Carcinoma Incidence in the United States Forecast Through 2030. J. Clin. Oncol. 2016, 34, 1787–1794. [Google Scholar] [CrossRef]
  5. Rawla, P.; Sunkara, T.; Muralidharan, P.; Raj, J.P. Update in global trends and aetiology of hepatocellular carcinoma. Współczesna Onkol. 2018, 22, 141–150. [Google Scholar] [CrossRef]
  6. Mittal, S.; Sada, Y.H.; El-Serag, H.B.; Kanwal, F.; Duan, Z.; Temple, S.; May, S.B.; Kramer, J.R.; Richardson, P.A.; Davila, J.A. Temporal trends of nonalcoholic fatty liver disease-related hepatocellular carcinoma in the veteran affairs population. Clin. Gastroenterol. Hepatol. 2015, 13, 594–601.e591. [Google Scholar] [CrossRef]
  7. Kallwitz, E.R.; Daviglus, M.L.; Allison, M.A.; Emory, K.T.; Zhao, L.; Kuniholm, M.H.; Chen, J.; Gouskova, N.; Pirzada, A.; Talavera, G.A.; et al. Prevalence of suspected nonalcoholic fatty liver disease in Hispanic/Latino individuals differs by heritage. Clin. Gastroenterol. Hepatol. 2015, 13, 569–576. [Google Scholar] [CrossRef]
  8. Chang, K.C.; Wu, Y.Y.; Hung, C.H.; Lu, S.N.; Lee, C.M.; Chiu, K.W.; Tsai, M.C.; Tseng, P.L.; Huang, C.M.; Cho, C.L.; et al. Clinical-guide risk prediction of hepatocellular carcinoma development in chronic hepatitis C patients after interferon-based therapy. Br. J. Cancer 2013, 109, 2481–2488. [Google Scholar] [CrossRef]
  9. Meredith, S.; Shiels, T.R.O.B. Recent Decline in Hepatocellular Carcinoma Rates in the United States. Gastroenterology 2020, 158, 1503–1505.e2. [Google Scholar]
  10. Medavaram, S.; Zhang, Y. Emerging therapies in advanced hepatocellular carcinoma. Exp. Hematol. Oncol. 2018, 7, 17. [Google Scholar] [CrossRef]
  11. McGlynn, K.A.; Petrick, J.L.; London, W.T. Global Epidemiology of Hepatocellular Carcinoma. Clin. Liver Dis. 2015, 19, 223–238. [Google Scholar] [CrossRef]
  12. Mittal, S.; El-Serag, H.B. Epidemiology of Hepatocellular Carcinoma. J. Clin. Gastroenterol. 2013, 47, S2–S6. [Google Scholar] [CrossRef]
  13. Raza, A. Hepatocellular carcinoma review: Current treatment, and evidence-based medicine. World J. Gastroenterol. 2014, 20, 4115. [Google Scholar] [CrossRef]
  14. Lewandowski, R.J.; Kulik, L.M.; Riaz, A.; Senthilnathan, S.; Mulcahy, M.F.; Ryu, R.K.; Ibrahim, S.M.; Sato, K.T.; Baker, T.; Miller, F.H.; et al. A Comparative Analysis of Transarterial Downstaging for Hepatocellular Carcinoma: Chemoembolization Versus Radioembolization. Am. J. Transpl. 2009, 9, 1920–1928. [Google Scholar] [CrossRef]
  15. Dimitroulis, D.; Damaskos, C.; Valsami, S.; Davakis, S.; Garmpis, N.; Spartalis, E.; Athanasiou, A.; Moris, D.; Sakellariou, S.; Kykalos, S.; et al. From diagnosis to treatment of hepatocellular carcinoma: An epidemic problem for both developed and developing world. World J. Gastroenterol. 2017, 23, 5282. [Google Scholar] [CrossRef]
  16. Daher, S.; Massarwa, M.; Benson, A.A.; Khoury, T. Current and Future Treatment of Hepatocellular Carcinoma: An Updated Comprehensive Review. J. Clin. Transl. Hepatol. 2018, 6, 69–78. [Google Scholar] [CrossRef]
  17. Lee, M.W.; Lim, H.K. Management of sub-centimeter recurrent hepatocellular carcinoma after curative treatment: Current status and future. World J. Gastroenterol. 2018, 24, 5215–5222. [Google Scholar] [CrossRef]
  18. Proneth, A.; Zeman, F.; Schlitt, H.J.; Schnitzbauer, A.A. Is resection or transplantation the ideal treatment in patients with hepatocellular carcinoma in cirrhosis if both are possible? A systematic review and metaanalysis. Ann. Surg. Oncol. 2014, 21, 3096–3107. [Google Scholar] [CrossRef]
  19. Schwartz, M. Liver transplantation in patients with hepatocellular carcinoma. Liver Transpl. 2004, 10, S81–S85. [Google Scholar] [CrossRef]
  20. Mazzaferro, V.; Bhoori, S.; Sposito, C.; Bongini, M.; Langer, M.; Miceli, R.; Mariani, L. Milan criteria in liver transplantation for hepatocellular carcinoma: An evidence-based analysis of 15 years of experience. Liver Transpl. 2011, 17 (Suppl. 2), S44–S57. [Google Scholar] [CrossRef]
  21. Higgins, H.; Berger, D.L. RFA for liver tumors: Does it really work? Oncologist 2006, 11, 801–808. [Google Scholar] [CrossRef]
  22. Salem, R.; Lewandowski, R.J.; Mulcahy, M.F.; Riaz, A.; Ryu, R.K.; Ibrahim, S.; Atassi, B.; Baker, T.; Gates, V.; Miller, F.H.; et al. Radioembolization for hepatocellular carcinoma using Yttrium-90 microspheres: A comprehensive report of long-term outcomes. Gastroenterology 2010, 138, 52–64. [Google Scholar] [CrossRef]
  23. Tohme, S.; Sukato, D.; Chen, H.W.; Amesur, N.; Zajko, A.B.; Humar, A.; Geller, D.A.; Marsh, J.W.; Tsung, A. Yttrium-90 radioembolization as a bridge to liver transplantation: A single-institution experience. J. Vasc. Interv. Radiol. 2013, 24, 1632–1638. [Google Scholar] [CrossRef]
  24. Kallini, J.R.; Gabr, A.; Salem, R.; Lewandowski, R.J. Transarterial Radioembolization with Yttrium-90 for the Treatment of Hepatocellular Carcinoma. Adv. Ther. 2016, 33, 699–714. [Google Scholar] [CrossRef]
  25. Mohamed, M.; Katz, A.W.; Tejani, M.A.; Sharma, A.K.; Kashyap, R.; Noel, M.S.; Qiu, H.; Hezel, A.F.; Ramaraju, G.A.; Dokus, M.K.; et al. Comparison of outcomes between SBRT, yttrium-90 radioembolization, transarterial chemoembolization, and radiofrequency ablation as bridge to transplant for hepatocellular carcinoma. Adv. Radiat. Oncol 2016, 1, 35–42. [Google Scholar] [CrossRef]
  26. Fidelman, N.; Kerlan, R.K. Transarterial Chemoembolization and90Y Radioembolization for Hepatocellular Carcinoma: Review of Current Applications Beyond Intermediate-Stage Disease. Am. J. Roentgenol. 2015, 205, 742–752. [Google Scholar] [CrossRef]
  27. Lee, J.J.X.; Choo, S.P. Locoregional therapy in hepatocullar carcinoma: When to start and when to stop and when to revisit. ESMO Open 2021, 6, 100129. [Google Scholar] [CrossRef]
  28. Ren, N.; Qin, S.; Ding, L.; Jia, E.; Xue, J. Comparison of Transarterial Y90 Radioembolization and Conventional Transarterial Chemoembolization in Hepatocarcinoma Patients: A Meta-analysis. Indian J. Pharm. Sci. 2020, 82, 76–81. [Google Scholar] [CrossRef]
  29. Yang, Y.; Si, T. Yttrium-90 transarterial radioembolization versus conventional transarterial chemoembolization for patients with hepatocellular carcinoma: A systematic review and meta-analysis. Cancer Biol. Med. 2018, 15, 299–310. [Google Scholar] [CrossRef]
  30. Vouche, M.; Habib, A.; Ward, T.J.; Kim, E.; Kulik, L.; Ganger, D.; Mulcahy, M.; Baker, T.; Abecassis, M.; Sato, K.T.; et al. Unresectable solitary hepatocellular carcinoma not amenable to radiofrequency ablation: Multicenter radiology-pathology correlation and survival of radiation segmentectomy. Hepatology 2014, 60, 192–201. [Google Scholar] [CrossRef]
  31. Kis, B.; El-Haddad, G.; Sheth, R.A.; Parikh, N.S.; Ganguli, S.; Shyn, P.B.; Choi, J.; Brown, K.T. Liver-Directed Therapies for Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. Cancer Control 2017, 24, 1073274817729244. [Google Scholar] [CrossRef]
  32. Viveiros, P.; Riaz, A.; Lewandowski, R.J.; Mahalingam, D. Current State of Liver-Directed Therapies and Combinatory Approaches with Systemic Therapy in Hepatocellular Carcinoma (HCC). Cancers 2019, 11, 1085. [Google Scholar] [CrossRef]
  33. Bahrambeigi, S.; Shafiei-Irannejad, V. Immune-mediated anti-tumor effects of metformin; targeting metabolic reprogramming of T cells as a new possible mechanism for anti-cancer effects of metformin. Biochem. Pharmacol. 2020, 174, 113787. [Google Scholar] [CrossRef]
  34. Guo, J.; Li, L.; Guo, B.; Liu, D.; Shi, J.; Wu, C.; Chen, J.; Zhang, X.; Wu, J. Mechanisms of resistance to chemotherapy and radiotherapy in hepatocellular carcinoma. Transl. Cancer Res. 2018, 7, 765–781. [Google Scholar] [CrossRef]
  35. Mukherjee, K.; Elsayed, M.; Choksi, E.; Loya, M.F.; Duszak, R.; Akce, M.; Majdalany, B.S.; Bercu, Z.L.; Cristescu, M.; Kokabi, N. Use of Metformin and Survival in Patients with Hepatocellular Carcinoma (HCC) Undergoing Liver Directed Therapy: Analysis of a Nationwide Cancer Registry. Cardiovasc. Interv. Radiol. 2023, 46, 870–879. [Google Scholar] [CrossRef]
  36. Facciorusso, A.; Del Prete, V.; Crucinio, N.; Muscatiello, N.; Carr, B.I.; Di Leo, A.; Barone, M. Angiotensin receptor blockers improve survival outcomes after radiofrequency ablation in hepatocarcinoma patients. J. Gastroenterol. Hepatol. 2015, 30, 1643–1650. [Google Scholar] [CrossRef]
  37. Yoshiji, H.; Kuriyama, S.; Fukui, H. Blockade of renin-angiotensin system in antifibrotic therapy. J. Gastroenterol. Hepatol. 2007, 22 (Suppl. 1), S93–S95. [Google Scholar] [CrossRef]
  38. Yoshiji, H.; Noguchi, R.; Toyohara, M.; Ikenaka, Y.; Kitade, M.; Kaji, K.; Yamazaki, M.; Yamao, J.; Mitoro, A.; Sawai, M.; et al. Combination of vitamin K2 and angiotensin-converting enzyme inhibitor ameliorates cumulative recurrence of hepatocellular carcinoma. J. Hepatol. 2009, 51, 315–321. [Google Scholar] [CrossRef]
  39. Yoshiji, H.; Noguchi, R.; Ikenaka, Y.; Kaji, K.; Aihara, Y.; Yamazaki, M.; Yamao, J.; Toyohara, M.; Mitoro, A.; Sawai, M.; et al. Combination of branched-chain amino acids and angiotensin-converting enzyme inhibitor suppresses the cumulative recurrence of hepatocellular carcinoma: A randomized control trial. Oncol. Rep. 2011, 26, 1547–1553. [Google Scholar] [CrossRef]
  40. Cabibbo, G.; Celsa, C.; Calvaruso, V.; Petta, S.; Cacciola, I.; Cannavò, M.R.; Madonia, S.; Rossi, M.; Magro, B.; Rini, F.; et al. Direct-acting antivirals after successful treatment of early hepatocellular carcinoma improve survival in HCV-cirrhotic patients. J. Hepatol. 2019, 71, 265–273. [Google Scholar] [CrossRef]
  41. Huang, G.; Lau, W.Y.; Wang, Z.G.; Pan, Z.Y.; Yuan, S.X.; Shen, F.; Zhou, W.P.; Wu, M.C. Antiviral therapy improves postoperative survival in patients with hepatocellular carcinoma: A randomized controlled trial. Ann. Surg. 2015, 261, 56–66. [Google Scholar] [CrossRef]
  42. Okamura, Y.; Sugiura, T.; Ito, T.; Yamamoto, Y.; Ashida, R.; Ohgi, K.; Uesaka, K. The Achievement of a Sustained Virological Response Either Before or After Hepatectomy Improves the Prognosis of Patients with Primary Hepatitis C Virus-Related Hepatocellular Carcinoma. Ann. Surg. Oncol. 2019, 26, 4566–4575. [Google Scholar] [CrossRef]
  43. Singal, A.G.; Rich, N.E.; Mehta, N.; Branch, A.D.; Pillai, A.; Hoteit, M.; Volk, M.; Odewole, M.; Scaglione, S.; Guy, J.; et al. Direct-Acting Antiviral Therapy for Hepatitis C Virus Infection Is Associated With Increased Survival in Patients With a History of Hepatocellular Carcinoma. Gastroenterology 2019, 157, 1253–1263.e1252. [Google Scholar] [CrossRef]
  44. Singal, A.K.; Salameh, H.; Kuo, Y.F.; Fontana, R.J. Meta-analysis: The impact of oral anti-viral agents on the incidence of hepatocellular carcinoma in chronic hepatitis B. Aliment. Pharmacol. Ther. 2013, 38, 98–106. [Google Scholar] [CrossRef]
  45. Wang, C.; Wang, X.; Su, Z.; Fei, H.; Liu, X.; Pan, Q. The novel mTOR inhibitor Torin-2 induces autophagy and downregulates the expression of UHRF1 to suppress hepatocarcinoma cell growth. Oncol. Rep. 2015, 34, 1708–1716. [Google Scholar] [CrossRef]
  46. Yin, J.; Li, N.; Han, Y.; Xue, J.; Deng, Y.; Shi, J.; Guo, W.; Zhang, H.; Wang, H.; Cheng, S.; et al. Effect of Antiviral Treatment With Nucleotide/Nucleoside Analogs on Postoperative Prognosis of Hepatitis B Virus–Related Hepatocellular Carcinoma: A Two-Stage Longitudinal Clinical Study. J. Clin. Oncol. 2013, 31, 3647–3655. [Google Scholar] [CrossRef]
  47. Yuan, P.; Chen, P.; Qian, Y. Evaluation of Antiviral Therapy Performed after Curative Therapy in Patients with HBV-Related Hepatocellular Carcinoma: An Updated Meta-Analysis. Can. J. Gastroenterol. Hepatol. 2016, 2016, 5234969. [Google Scholar] [CrossRef]
  48. Wang, G.; Duan, Z. Guidelines for Prevention and Treatment of Chronic Hepatitis B. J. Clin. Transl. Hepatol. 2021, 9, 769–791. [Google Scholar] [CrossRef]
  49. Zhong, J.H.; Mo, X.S.; Xiang, B.D.; Yuan, W.P.; Jiang, J.F.; Xie, G.S.; Li, L.Q. Postoperative use of the chemopreventive vitamin K2 analog in patients with hepatocellular carcinoma. PLoS ONE 2013, 8, e58082. [Google Scholar] [CrossRef]
  50. Hitomi, M.; Nonomura, T.; Yokoyama, F.; Yoshiji, H.; Ogawa, M.; Nakai, S.; Deguchi, A.; Masaki, T.; Inoue, H.; Kimura, Y.; et al. In vitro and in vivo antitumor effects of vitamin K5 on hepatocellular carcinoma. Int. J. Oncol. 2005, 26, 1337–1344. [Google Scholar] [CrossRef]
  51. Hitomi, M.; Yokoyama, F.; Kita, Y.; Nonomura, T.; Masaki, T.; Yoshiji, H.; Inoue, H.; Kinekawa, F.; Kurokohchi, K.; Uchida, N.; et al. Antitumor effects of vitamins K1, K2 and K3 on hepatocellular carcinoma in vitro and in vivo. Int. J. Oncol. 2005, 26, 713–720. [Google Scholar] [CrossRef]
  52. Hotta, N.; Ayada, M.; Sato, K.; Ishikawa, T.; Okumura, A.; Matsumoto, E.; Ohashi, T.; Kakumu, S. Effect of vitamin K2 on the recurrence in patients with hepatocellular carcinoma. Hepatogastroenterology 2007, 54, 2073–2077. [Google Scholar]
  53. Kakizaki, S.; Sohara, N.; Sato, K.; Suzuki, H.; Yanagisawa, M.; Nakajima, H.; Takagi, H.; Naganuma, A.; Otsuka, T.; Takahashi, H.; et al. Preventive effects of vitamin K on recurrent disease in patients with hepatocellular carcinoma arising from hepatitis C viral infection. J. Gastroenterol. Hepatol. 2007, 22, 518–522. [Google Scholar] [CrossRef]
  54. Mizuta, T.; Ozaki, I.; Eguchi, Y.; Yasutake, T.; Kawazoe, S.; Fujimoto, K.; Yamamoto, K. The effect of menatetrenone, a vitamin K2 analog, on disease recurrence and survival in patients with hepatocellular carcinoma after curative treatment: A pilot study. Cancer 2006, 106, 867–872. [Google Scholar] [CrossRef]
  55. Yoshida, H.; Shiratori, Y.; Kudo, M.; Shiina, S.; Mizuta, T.; Kojiro, M.; Yamamoto, K.; Koike, Y.; Saito, K.; Koyanagi, N.; et al. Effect of vitamin K2 on the recurrence of hepatocellular carcinoma. Hepatology 2011, 54, 532–540. [Google Scholar] [CrossRef]
  56. Xue, L.; Chen, F.; Yue, F.; Camacho, L.; Kothapalli, S.; Wei, G.; Huang, S.; Mo, Q.; Ma, F.; Li, Y.; et al. Metformin and an insulin/IGF-1 receptor inhibitor are synergistic in blocking growth of triple-negative breast cancer. Breast Cancer Res. Treat. 2021, 185, 73–84. [Google Scholar] [CrossRef]
  57. Wang, Y.; An, H.; Liu, T.; Qin, C.; Sesaki, H.; Guo, S.; Radovick, S.; Hussain, M.; Maheshwari, A.; Wondisford, F.E.; et al. Metformin Improves Mitochondrial Respiratory Activity through Activation of AMPK. Cell Rep. 2019, 29, 1511–1523.e1515. [Google Scholar] [CrossRef]
  58. Leprivier, G.; Rotblat, B. How does mTOR sense glucose starvation? AMPK is the usual suspect. Cell Death Discov. 2020, 6, 27. [Google Scholar] [CrossRef]
  59. Li, L.; Wang, T.; Hu, M.; Zhang, Y.; Chen, H.; Xu, L. Metformin Overcomes Acquired Resistance to EGFR TKIs in EGFR-Mutant Lung Cancer via AMPK/ERK/NF-κB Signaling Pathway. Front. Oncol. 2020, 10, 1605. [Google Scholar] [CrossRef]
  60. Lv, Z.; Guo, Y. Metformin and Its Benefits for Various Diseases. Front. Endocrinol. 2020, 11, 191. [Google Scholar] [CrossRef]
  61. Zhou, Z.; Liu, Y.; Song, W.; Jiang, X.; Deng, Z.; Xiong, W.; Shen, J. Metabolic reprogramming mediated PD-L1 depression and hypoxia reversion to reactivate tumor therapy. J. Control. Release 2022, 352, 793–812. [Google Scholar] [CrossRef]
  62. Evans, J.M.M.; Donnelly, L.A.; Emslie-Smith, A.M.; Alessi, D.R.; Morris, A.D. Metformin and reduced risk of cancer in diabetic patients. BMJ 2005, 330, 1304–1305. [Google Scholar] [CrossRef]
  63. Bowker, S.L.; Majumdar, S.R.; Veugelers, P.; Johnson, J.A. Increased Cancer-Related Mortality for Patients With Type 2 Diabetes Who Use Sulfonylureas or Insulin. Diabetes Care 2006, 29, 254–258. [Google Scholar] [CrossRef]
  64. Donadon, V.; Balbi, M.; Ghersetti, M.; Grazioli, S.; Perciaccante, A.; Della Valentina, G.; Gardenal, R.; Dal Mas, M.; Casarin, P.; Zanette, G.; et al. Antidiabetic therapy and increased risk of hepatocellular carcinoma in chronic liver disease. World J. Gastroenterol. 2009, 15, 2506–2511. [Google Scholar] [CrossRef]
  65. Hassan, M.M.; Curley, S.A.; Li, D.; Kaseb, A.; Davila, M.; Abdalla, E.K.; Javle, M.; Moghazy, D.M.; Lozano, R.D.; Abbruzzese, J.L.; et al. Association of diabetes duration and diabetes treatment with the risk of hepatocellular carcinoma. Cancer 2010, 116, 1938–1946. [Google Scholar] [CrossRef]
  66. Landman, G.W.; van Hateren, K.J.; Kleefstra, N.; Groenier, K.H.; Gans, R.O.; Bilo, H.J. The relationship between glycaemic control and mortality in patients with type 2 diabetes in general practice (ZODIAC-11). Br. J. Gen. Pract. 2010, 60, 172–175. [Google Scholar] [CrossRef]
  67. Lee, M.S.; Hsu, C.C.; Wahlqvist, M.L.; Tsai, H.N.; Chang, Y.H.; Huang, Y.C. Type 2 diabetes increases and metformin reduces total, colorectal, liver and pancreatic cancer incidences in Taiwanese: A representative population prospective cohort study of 800,000 individuals. BMC Cancer 2011, 11, 20. [Google Scholar] [CrossRef]
  68. Lai, S.W.; Chen, P.C.; Liao, K.F.; Muo, C.H.; Lin, C.C.; Sung, F.C. Risk of hepatocellular carcinoma in diabetic patients and risk reduction associated with anti-diabetic therapy: A population-based cohort study. Am. J. Gastroenterol. 2012, 107, 46–52. [Google Scholar] [CrossRef]
  69. Deperalta, D.K.; Wei, L.; Ghoshal, S.; Schmidt, B.; Lauwers, G.Y.; Lanuti, M.; Chung, R.T.; Tanabe, K.K.; Fuchs, B.C. Metformin prevents hepatocellular carcinoma development by suppressing hepatic progenitor cell activation in a rat model of cirrhosis. Cancer 2016, 122, 1216–1227. [Google Scholar] [CrossRef]
  70. Kasmari, A.J.; Welch, A.; Liu, G.; Leslie, D.; McGarrity, T.; Riley, T. Independent of Cirrhosis, Hepatocellular Carcinoma Risk Is Increased with Diabetes and Metabolic Syndrome. Am. J. Med. 2017, 130, 746.e1–746.e7. [Google Scholar] [CrossRef]
  71. Yu, H.; Yin, L.; Jiang, X.; Sun, X.; Wu, J.; Tian, H.; Gao, X.; He, X. Effect of Metformin on Cancer Risk and Treatment Outcome of Prostate Cancer: A Meta-Analysis of Epidemiological Observational Studies. PLoS ONE 2014, 9, e116327. [Google Scholar] [CrossRef]
  72. Stopsack, K.H.; Ziehr, D.R.; Rider, J.R.; Giovannucci, E.L. Metformin and prostate cancer mortality: A meta-analysis. Cancer Causes Control 2016, 27, 105–113. [Google Scholar] [CrossRef]
  73. Nevadunsky, N.S.; Van Arsdale, A.; Strickler, H.D.; Moadel, A.; Kaur, G.; Frimer, M.; Conroy, E.; Goldberg, G.L.; Einstein, M.H. Metformin use and endometrial cancer survival. Gynecol. Oncol. 2014, 132, 236–240. [Google Scholar] [CrossRef]
  74. Tarhini, Z.; Manceur, K.; Magne, J.; Mathonnet, M.; Jost, J.; Christou, N. The effect of metformin on the survival of colorectal cancer patients with type 2 diabetes mellitus. Sci. Rep. 2022, 12, 12374. [Google Scholar] [CrossRef]
  75. Rêgo, D.F.; Pavan, L.M.; Elias, S.T.; De Luca Canto, G.; Guerra, E.N. Effects of metformin on head and neck cancer: A systematic review. Oral Oncol. 2015, 51, 416–422. [Google Scholar] [CrossRef]
  76. Shankaraiah, R.C.; Callegari, E.; Guerriero, P.; Rimessi, A.; Pinton, P.; Gramantieri, L.; Silini, E.M.; Sabbioni, S.; Negrini, M. Metformin prevents liver tumourigenesis by attenuating fibrosis in a transgenic mouse model of hepatocellular carcinoma. Oncogene 2019, 38, 7035–7045. [Google Scholar] [CrossRef]
  77. Cai, X.; Hu, X.; Cai, B.; Wang, Q.; Li, Y.; Tan, X.; Hu, H.; Chen, X.; Huang, J.; Cheng, J.; et al. Metformin suppresses hepatocellular carcinoma cell growth through induction of cell cycle G1/G0 phase arrest and p21CIP and p27KIP expression and downregulation of cyclin D1 in vitro and in vivo. Oncol. Rep. 2013, 30, 2449–2457. [Google Scholar] [CrossRef]
  78. Bhalla, K.; Hwang, B.J.; Dewi, R.E.; Twaddel, W.; Goloubeva, O.G.; Wong, K.-K.; Saxena, N.K.; Biswal, S.; Girnun, G.D. Metformin Prevents Liver Tumorigenesis by Inhibiting Pathways Driving Hepatic Lipogenesis. Cancer Prev. Res. 2012, 5, 544–552. [Google Scholar] [CrossRef]
  79. Guo, W.J.; Yu, E.X.; Liu, L.M.; Li, J.; Chen, Z.; Lin, J.H.; Meng, Z.Q.; Feng, Y. Comparison between chemoembolization combined with radiotherapy and chemoembolization alone for large hepatocellular carcinoma. World J. Gastroenterol. 2003, 9, 1697–1701. [Google Scholar] [CrossRef]
  80. Facciuto, M.E.; Singh, M.K.; Rochon, C.; Sharma, J.; Gimenez, C.; Katta, U.; Moorthy, C.R.; Bentley-Hibbert, S.; Rodriguez-Davalos, M.; Wolf, D.C. Stereotactic body radiation therapy in hepatocellular carcinoma and cirrhosis: Evaluation of radiological and pathological response. J. Surg. Oncol. 2012, 105, 692–698. [Google Scholar] [CrossRef]
  81. Hozo, S.P.; Djulbegovic, B.; Hozo, I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med. Res. Methodol. 2005, 5, 13. [Google Scholar] [CrossRef]
  82. Chen, T.M.; Lin, C.C.; Huang, P.T.; Wen, C.F. Metformin associated with lower mortality in diabetic patients with early stage hepatocellular carcinoma after radiofrequency ablation. J. Gastroenterol. Hepatol. 2011, 26, 858–865. [Google Scholar] [CrossRef]
  83. Chen, M.L.; Wu, C.X.; Zhang, J.B.; Zhang, H.; Sun, Y.D.; Tian, S.L.; Han, J.J. Transarterial chemoembolization combined with metformin improves the prognosis of hepatocellular carcinoma patients with type 2 diabetes. Front. Endocrinol. 2022, 13, 996228. [Google Scholar] [CrossRef]
  84. Elsayed, M.; Wagstaff, W.; Behbahani, K.; Villalobos, A.; Bercu, Z.; Majdalany, B.S.; Akce, M.; Schuster, D.M.; Mao, H.; Kokabi, N. Improved Tumor Response in Patients on Metformin Undergoing Yttrium-90 Radioembolization Segmentectomy for Hepatocellular Carcinoma. Cardiovasc. Interv. Radiol. 2021, 44, 1937–1944. [Google Scholar] [CrossRef]
  85. Jang, W.I.; Kim, M.S.; Lim, J.S.; Yoo, H.J.; Seo, Y.S.; Han, C.J.; Park, S.C.; Kay, C.S.; Kim, M.; Jang, H.S.; et al. Survival Advantage Associated with Metformin Usage in Hepatocellular Carcinoma Patients Receiving Radiotherapy: A Propensity Score Matching Analysis. Anticancer Res. 2015, 35, 5047–5054. [Google Scholar]
  86. Jung, W.J.; Jang, S.; Choi, W.J.; Park, J.; Choi, G.H.; Jang, E.S.; Jeong, S.H.; Choi, W.S.; Lee, J.H.; Yoon, C.J.; et al. Metformin administration is associated with enhanced response to transarterial chemoembolization for hepatocellular carcinoma in type 2 diabetes patients. Sci. Rep. 2022, 12, 14482. [Google Scholar] [CrossRef]
  87. Kim, E.H.; Kim, M.S.; Cho, C.K.; Jung, W.G.; Jeong, Y.K.; Jeong, J.H. Low and high linear energy transfer radiation sensitization of HCC cells by metformin. J. Radiat. Res. 2014, 55, 432–442. [Google Scholar] [CrossRef]
  88. Liu, J.; Hou, M.; Yuan, T.; Yi, G.; Zhang, S.; Shao, X.; Chen, J.; Jia, X.; He, Z. Enhanced cytotoxic effect of low doses of metformin combined with ionizing radiation on hepatoma cells via ATP deprivation and inhibition of DNA repair. Oncol. Rep. 2012, 28, 1406–1412. [Google Scholar] [CrossRef]
  89. Tian, Y.; Tang, B.; Wang, C.; Sun, D.; Zhang, R.; Luo, N.; Han, Z.; Liang, R.; Gao, Z.; Wang, L. Metformin mediates resensitivity to 5-fluorouracil in hepatocellular carcinoma via the suppression of YAP. Oncotarget 2016, 7, 46230–46241. [Google Scholar] [CrossRef]
  90. Zhang, Q.; Kong, J.; Dong, S.; Xu, W.; Sun, W. Metformin exhibits the anti-proliferation and anti-invasion effects in hepatocellular carcinoma cells after insufficient radiofrequency ablation. Cancer Cell Int. 2017, 17, 48. [Google Scholar] [CrossRef]
  91. Zhang, K.F.; Wang, J.; Guo, J.; Huang, Y.Y.; Huang, T.R. Metformin enhances radiosensitivity in hepatocellular carcinoma by inhibition of specificity protein 1 and epithelial-to-mesenchymal transition. J. Cancer Res. Ther. 2019, 15, 1603–1610. [Google Scholar] [CrossRef]
  92. Xin, H.W.; Ambe, C.M.; Miller, T.C.; Chen, J.Q.; Wiegand, G.W.; Anderson, A.J.; Ray, S.; Mullinax, J.E.; Hari, D.M.; Koizumi, T.; et al. Liver Label Retaining Cancer Cells Are Relatively Resistant to the Reported Anti-Cancer Stem Cell Drug Metformin. J. Cancer 2016, 7, 1142–1151. [Google Scholar] [CrossRef]
  93. Kim, H.S.; El-Serag, H.B. The Epidemiology of Hepatocellular Carcinoma in the USA. Curr. Gastroenterol. Rep. 2019, 21, 17. [Google Scholar] [CrossRef]
  94. Li, J.; Hernanda, P.Y.; Bramer, W.M.; Peppelenbosch, M.P.; van Luijk, J.; Pan, Q. Anti-tumor effects of metformin in animal models of hepatocellular carcinoma: A systematic review and meta-analysis. PLoS ONE 2015, 10, e0127967. [Google Scholar] [CrossRef]
  95. Coyle, C.; Cafferty, F.H.; Vale, C.; Langley, R.E. Metformin as an adjuvant treatment for cancer: A systematic review and meta-analysis. Ann. Oncol. 2016, 27, 2184–2195. [Google Scholar] [CrossRef]
  96. Ng, C.W.; Jiang, A.A.; Toh, E.M.S.; Ng, C.H.; Ong, Z.H.; Peng, S.; Tham, H.Y.; Sundar, R.; Chong, C.S.; Khoo, C.M. Metformin and colorectal cancer: A systematic review, meta-analysis and meta-regression. Int. J. Color. Dis. 2020, 35, 1501–1512. [Google Scholar] [CrossRef]
  97. Zhang, K.; Bai, P.; Dai, H.; Deng, Z. Metformin and risk of cancer among patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Prim. Care Diabetes 2021, 15, 52–58. [Google Scholar] [CrossRef]
  98. Chen, Y.H.; Wang, P.H.; Chen, P.N.; Yang, S.F.; Hsiao, Y.H. Molecular and Cellular Mechanisms of Metformin in Cervical Cancer. Cancers 2021, 13, 2545. [Google Scholar] [CrossRef]
  99. Tan, X.; Li, Y.; Hou, Z.; Zhang, M.; Li, L.; Wei, J. Combination therapy with PD-1 inhibition plus rapamycin and metformin enhances anti-tumor efficacy in triple negative breast cancer. Exp. Cell Res. 2023, 429, 113647. [Google Scholar] [CrossRef]
  100. Zhou, Z.; Liu, Y.; Jiang, X.; Zheng, C.; Luo, W.; Xiang, X.; Qi, X.; Shen, J. Metformin modified chitosan as a multi-functional adjuvant to enhance cisplatin-based tumor chemotherapy efficacy. Int. J. Biol. Macromol. 2023, 224, 797–809. [Google Scholar] [CrossRef]
  101. Cheaib, B.; Auguste, A.; Leary, A. The PI3K/Akt/mTOR pathway in ovarian cancer: Therapeutic opportunities and challenges. Chin. J. Cancer 2015, 34, 4–16. [Google Scholar] [CrossRef] [PubMed]
  102. Zhou, W.; Marcus, A.I.; Vertino, P.M. Dysregulation of mTOR activity through LKB1 inactivation. Chin. J. Cancer 2013, 32, 427–433. [Google Scholar] [CrossRef]
  103. Karuman, P.; Gozani, O.; Odze, R.D.; Zhou, X.C.; Zhu, H.; Shaw, R.; Brien, T.P.; Bozzuto, C.D.; Ooi, D.; Cantley, L.C.; et al. The Peutz-Jegher gene product LKB1 is a mediator of p53-dependent cell death. Mol. Cell 2001, 7, 1307–1319. [Google Scholar] [CrossRef] [PubMed]
  104. Algire, C.; Amrein, L.; Zakikhani, M.; Panasci, L.; Pollak, M. Metformin blocks the stimulative effect of a high-energy diet on colon carcinoma growth in vivo and is associated with reduced expression of fatty acid synthase. Endocr. Relat. Cancer 2010, 17, 351–360. [Google Scholar] [CrossRef] [PubMed]
  105. Hayashi, T.; Yamashita, T.; Okada, H.; Oishi, N.; Sunagozaka, H.; Nio, K.; Hayashi, T.; Hara, Y.; Asahina, Y.; Yoshida, M.; et al. A Novel mTOR Inhibitor; Anthracimycin for the Treatment of Human Hepatocellular Carcinoma. Anticancer Res. 2017, 37, 3397–3403. [Google Scholar] [CrossRef]
  106. Snima, K.S.; Jayakumar, R.; Unnikrishnan, A.G.; Nair, S.V.; Lakshmanan, V.K. O-carboxymethyl chitosan nanoparticles for metformin delivery to pancreatic cancer cells. Carbohydr. Polym. 2012, 89, 1003–1007. [Google Scholar] [CrossRef]
  107. Verdura, S.; Cuyàs, E.; Martin-Castillo, B.; Menendez, J.A. Metformin as an archetype immuno-metabolic adjuvant for cancer immunotherapy. OncoImmunology 2019, 8, e1633235. [Google Scholar] [CrossRef]
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA 2020) diagram.
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA 2020) diagram.
Cancers 15 04538 g001
Table 1. Demographics of included preclinical and clinical studies.
Table 1. Demographics of included preclinical and clinical studies.
Author, YearStudy TypeLocation of StudyNo. of Patients 1Sex, M:F RatioMean Age (Years)Type of HCC Intervention 2Control Group (# of Patients) 3Experimental Group (# of Patients) 3
Chen 2011 [82]Human clinicalTaiwan13584:5163.0 ± 9.7LDTRFA (114)RFA + MET (21)
Chen 2022 [83]Human clinicalChina123108:15-LDTTACE (73)TACE + MET (50)
Elsayed 2021 [84]Human clinicalUSA10372:3464.2 ± 8.9LDTY-90 RS (93)Y-90 RS + MET (19)
Jang 2015 [85]Human clinicalKorea21757:160-EBRTSBRT or HypoRT (198)SBRT or HypoRT + MET (10)
Jung 2022 [86]Human, clinicalSouth Korea16475:8969.0 ± 16.4LDTTACE (73)TACE + MET (91)
Kim 2014 [87]Human pre-clinicalSouth Korea---EBRTy-ray radiation and neutron radiationy-ray radiation/neutron radiation + MET
Liu 2012 [88]Human pre-clinicalChina---EBRTIonizing radiationIonizing radiation + MET
Tian 2016 [89]Human pre-clinical/clinicalChina6311:52-LDTTACE (30)TACE + MET (33)
Zhang 2017 [90]Human pre-clinicalChina---LDTRFARFA + MET
Zhang 2019 [91]Human pre-clinicalChina---EBRTGamma ray irradiationGamma ray irradiation + MET
1 Pre-clinical studies did not have patients; 2 LDT = liver-directed therapy, EBR = external-beam radiation therapy; 3 TACE = transarterial chemoembolization, MET = metformin, RFA = radiofrequency ablation, RS = radiosensitization, SBRT = stereotactic body radiotherapy, HypoRT = hypofractioned radiotherapy.
Table 2. Outcomes of human clinical studies in control and experimental groups.
Table 2. Outcomes of human clinical studies in control and experimental groups.
StudyOverall Survival (Control)Overall Survival (Experimental)Progression-Free Survival (Control)Progression Free Survival (Experimental)Results of Study 1Metformin Dosage
Jung 2022 [86]--74 months79 monthsMetformin use associated with higher ORR, lower LTR, better IR, and lower recurrence-
Elsayed 2021 [84]39.7 months33.7 months--MET + Y-90 RS had greater reduction in TTD, number of lesions, and % TTD-
Jang 2015 [85]37% 2 years76% 2 years16% 2 years46% 2 yearsUse of metformin in patients receiving radiotherapy was associated with higher OS1000 mg/day
Chen 2011 [82]93.9% 1 year; 80.2% 3 years; 64.7% 5 years95.0% 1 year; 69.2% 3 years; 60.5% 5 years74.5% 1 year; 44.8% 3 years; 26.2% 5 years95% 1 year; 69.2% 3 years; 60.5% 5 yearsMetformin users among diabetic patients with HCC undergoing RFA had favorable overall survival compared with patients without metformin750 mg/day
Chen 2022 [83]32 months42 months7 months12 monthsMetformin increased long-term survival time and significantly prevented recurrence after TACE2 g/day
Tian 2016 [89]75 months79 months--Metformin + TACE combination resulted in increased OS-
1 ORR = overall response rate, LTR = local tumor recurrence, IR = initial response, TTD = total tumor diameter, RFA = radiofrequency ablation, HCC = hepatocellular carcinoma, TACE = transarterial chemoembolization, OS = overall survival, PFS = progression-free survival.
Table 3. Outcomes of Human Pre-Clinical Studies in Control and Experimental Groups.
Table 3. Outcomes of Human Pre-Clinical Studies in Control and Experimental Groups.
StudyCell Type UsedResults of Study 1Metformin Dosage
Zhang 2017 [90]HepG2 and SMMC7721MET + RFA inhibited proliferation, migration, and invasion of Hep G2 cells after insufficient RFA. MET also blocked growth of HepG2 cells0.5 mg/mL
Zhang 2019 [91]SMMC-7721Metformin demonstrated enhanced radiosensitivity and inhibition of EMT in HCC cells.0, 1, 5, 10, and 20 mM
Tian 2016 [89]Bel7402 and Bel7402-5 fluorouracil5-FU + MET increased chemotherapeutic sensitivity, decreased cell proliferation, increased apoptosis, and arrested cell cycle at G0/G12 mg/kg
Liu 2012 [88]HepG2 and Bel-7402 cellsCombination of Met + ionizing radiation inhibited cell survival, cell cycle progression, and increased DNA damage more than monotherapies-
Kim 2014 [87]Huh7, HepG2 and Hep3BCombination of y-ray radiation + MET enhanced radiation efficacy, increased apoptosis, increased DNA damage, and stopped G2/M cell cycle in HCC cells-
Xin 2016 [92]PLC/PRF/5, SK-Hep-1, Huh7MET increased effects of Sorafenib and significantly decreased viability/proliferation of HCC cells200 μM
1 MET = metformin, HCC = hepatocellular carcinoma, RFA = radiofrequency ablation, EMT = epithelial-to-mesenchymal transition, 5-FU = 5-fluorouracil.
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

Choksi, E.J.; Elsayed, M.; Kokabi, N. Antitumor Activity of Metformin Combined with Locoregional Therapy for Liver Cancer: Evidence and Future Directions. Cancers 2023, 15, 4538. https://doi.org/10.3390/cancers15184538

AMA Style

Choksi EJ, Elsayed M, Kokabi N. Antitumor Activity of Metformin Combined with Locoregional Therapy for Liver Cancer: Evidence and Future Directions. Cancers. 2023; 15(18):4538. https://doi.org/10.3390/cancers15184538

Chicago/Turabian Style

Choksi, Eshani J., Mohammad Elsayed, and Nima Kokabi. 2023. "Antitumor Activity of Metformin Combined with Locoregional Therapy for Liver Cancer: Evidence and Future Directions" Cancers 15, no. 18: 4538. https://doi.org/10.3390/cancers15184538

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

Article Metrics

Back to TopTop