Download PDF
Original Article  |  Open Access  |  14 Jul 2015

Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma

Views: 8972 |  Downloads: 917 |  Cited:   0
Hepatoma Res 2015;1:92-6.
Author Information
Article Notes
Cite This Article

Abstract

Aim: Used as a palliative therapy for unresectable liver cancer, radiofrequency ablation (RFA) is associated with the induction of immunological responses. Here, we show strong evidence of tumor-specific peripheral blood mononuclear cells (PBMCs) 12 months after RFA.

Methods: Three patients with colorectal cancer (CRC) metastases to the liver and two patients with primary hepatocellular carcinoma (HCC) were enrolled in this study. PBMC, isolated 12 months after RFA, were stimulated with normal and tumor tissue lysate. Interferon gamma secretion was evaluated by flow cytometry and indirectly, by luciferase assay for adenylate kinase activity in PBMC-stimulated lysates of target cells. Baseline data were detected before RFA and 4 weeks after treatment.

Results: Two CRC patients and one HCC patient had recurrence-free survival. One patient with CRC developed secondary metastases; one patient with HCC developed a local recurrence. Recurrence-free patients showed a significantly higher cytolytic activity of PBMC against matched tumor cells 12 months after RFA treatment. Interestingly, patients with malignant recurrence showed a decreased cytolytic activity.

Conclusion: RFA seems to overcome immune-tolerance toward tumor antigens and/or presents new tumor antigens. Patients seem to benefit from a prolonged increase in cytolytic activity. The immune-modulatory effects of RFA need further investigations in multimodality anticancer therapies.

Keywords

Liver cancer, radiofrequency ablation, immune response, interferon gamma, peripheral blood mononuclear cells

Introduction

Surgical resection is still the gold standard for the treatment of hepatocellular carcinoma (HCC) and liver metastases of colorectal cancer (CRC). However, more than 75% of these patients are elected as unresectable due to the volume and localization of the tumor. Radiofrequency ablation (RFA) is a common therapy option for unresectable liver tumors.[1-4] It was shown that RFA and laser-induced thermotherapy (LiTT) can achieve, in selected patients, a survival prolongation time comparable to surgical resection.

An earlier study suggested that RFA has adjunctive immune-modulatory side effects.[5] By using the VX 2 hepatoma model in rabbits, we showed that RFA can induce a strong mononuclear infiltration around the implanted tumor. More recently, we demonstrated a marked tumor-specific peripheral T cell response in RFA-treated vs. untreated rabbits with the VX 2 hepatoma.[6,7] We further investigated whether this strong immune response to RFA can be observed in humans. We observed that significantly elevated levels of CD8+ T cells appeared 4 weeks after RFA, and this effect lasted at least up to 8 weeks after RFA. There was also significant cytolytic activity of isolated peripheral blood mononuclear cells (PBMCs) 8 weeks after RFA.[8]

The aim of this study was to investigate the immunological features of HCC and CRC metastases patients 12 months after RFA.

Methods

The study was approved by the Ethical Committee of the University of Erlangen-Nuremberg (Ethikkomitee der Universität Erlangen Nürnberg) and performed according to the declaration of Helsinki. All treated HCC and CRC metastases were confirmed histologically prior to therapy.

Selection and description of participants

All enrolled patients took part in a prior trial in which baseline data before RFA treatment were recorded according to the present protocol.

Patients with up to 3 tumor nodules within the liver, with a maximum diameter of 6 cm per lesion, were enrolled in the study. Prior local ablative therapy (LiTT, RFA, ethanol injection) or prior chemo-embolization of the malignant liver tumor was an exclusion criterion. The possibility of curative treatment by resection had to be ruled out. Therefore, all cases were discussed in our tumor conference, including gastroenterologists, oncologists, surgeons, and radiologists.

Patients with at least one of the following findings were also excluded: Karnofsky index < 60, thrombocytes < 50,000/μL, prothrombin activity < 50%, partial thromboplastin time > 80 s. No transfusion of platelets or fresh frozen plasma was performed. Informed consent was obtained from every patient no later than 24 h before treatment.

The size and number of tumor nodules were determined by ultrasonography and by computed tomography (CT) (dynamic spiral CT with intravenous application of contrast medium) prior to RFA.

Five consecutive male patients with 8 tumor nodules in total (CRC patients with 2 nodules each and HCC patients with 1 nodule) who met the inclusion criteria were enrolled. Mean patient age was 64 years (range: 59-74 years). Three patients suffered from CRC metastases to the liver while 2 suffering from HCC.

Radiofrequency ablation technique

The whole procedure was performed under ultrasound guidance (Elegra Advanced®, Siemens, Erlangen, Germany) under sterile conditions. The proposed puncture site was infiltrated with a local anesthetic (2% mepivacaine hydrochloride) and the perfused radio-frequency (HF) needle (Integra, Rätingen, Germany) advanced into the tumor. Midazolam (0.5-5 mg) and/or pethidine (25-100 mg) were administered intravenously as necessary. Patients were monitored by pulse oximetry during the whole procedure.

Two mm (14 G) diameter RFA needle applicators and 15 mm active electrode with microbores were used. During HF application (40 W power output) the RFA needle was continuously perfused with isotonic saline via the bore holes. RF energy was delivered by a computer-assisted radiofrequency generator (Elektrotom 106 HF®, Integra, Rätingen, Germany) and continuous perfusion of the RFA needle was secured by a syringe pump (Pilot C, Fresenius Medical Care, Alzenau, Germany) linked to the RF generator. Perfusion was adjusted according to impedance by means of an electronic interface between generator and perfusor, automatically increasing in response to a rise in impedance (> 400 Ohm). The RF energy was applied for 10-15 min at each needle position, leading to a coagulation zone of 30-35 mm in diameter. Tumors larger than 20 mm were targeted using different applicator positions to create overlapping coagulation zones, in order to treat the entire lesion with a safety margin > 5 mm. Larger tumors were treated with up to 3 simultaneous needle insertions arranged in a triangle (2-4 cm) or square (for larger tumors).

Interferon gamma secretion assay and lymphocyte staining

Heparinized blood (LI-Heparin 10 mL) was collected 12 months after RFA. The samples were stored at 4 °C and tests performed within 12 h after sampling. A liver biopsy of normal and tumor tissue was collected from every patient directly before RFA and stored at -20 °C.

Tissue-lysates were freshly prepared in cold phosphate buffer (50 mmol/L, pH 7.2) using a glass homogenizer as described.[6] The suspension was filtered with a filter tip (pore size 1.2 mm) to adjust the fragment size to < 1.2 mm. The protein concentration was measured spectrophotometrically using the Bradford assay, and adjusted to 1 mg/mL.

Autologous test antigens (normal and tumor lysate, 12.5 mg) were added to 250 μL heparinized blood and cultured in a 15 mL conical polypropylene tube for 16 h at 37 °C under 5% CO2. A negative control without the addition of antigen lysate was included, while staphylococcal enterotoxin B served as positive control antigen. Thereafter, the samples were put on ice and washed with ice cold washing solution [phosphate buffer saline containing 0.5% bovine serum albumin and 2 mmol/L ethylenediaminetetraacetic acid (EDTA), pH 7.4] and the cell suspension centrifuged at 300 g for 10 min at 4 °C. The cell pellet was resuspended with 80 μL ice cold culture medium (RPMI1640 containing 10% human AB serum). "Catch" reagent (20 μL) containing a bivalent CD45 capture and interferon gamma (IFNγ) binding antibody (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) were added; the suspension was kept for 5 min on ice and 5 mL medium was added before incubation at 37 °C in closed roller tubes for 45 min. Thereafter, 20 μL phycoerythrin-conjugated antibody against IFNγ (1:5 dilution ratio) as well as 10 μL fluoresceine-thiocyanate conjugated anti-CD8 antibody (1:400 dilution ratio) (both from Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) were added to the cooled and washed cell suspension and incubated for 10 min on ice. Erythrocyte lysis buffer (5 mL), containing 0.155 mol/L NH4Cl, 10 mmol/L KHCO3 and 0.1 mmol/L EDTA diluted 1:10, was added for 10 min and the cell suspension centrifuged at 300 g for 10 min. The cell pellet was resuspended in 500 μL ice cold washing buffer and immediately analyzed by flow cytometry (FACS Calibur, BD Biosciences, Heidelberg, Germany) after addition of 0.25 mg propidium iodide in 5 μL distilled water. FACS data were analyzed using software Win MDI Version 2.8 (Scripps Research Institute, La Jolla, CA, USA).

Cytotoxicity assay

Cytolytic activity of T cells was measured by adenylate kinase (AK) release assay. Cells (104) were incubated with 1,000 effector cells in a final volume of 200 μL growth medium with fetal calf serum in round-bottom 96-well microtiter plates. After incubation for 4 h at 37 °C, 100 μL of supernatant was harvested and stored at -20 °C for further analysis.

The human HCC cell line HepG2 and the human CRC cell line CaCO2 served as target cells for T cells isolated from patients with HCC and CRC metastases, respectively. All cell lines were human leukocyte antigen matched (ABO-system) and tested previously. HepG2 (ACC-180) and CaCO2 (ACC169) cells were purchased from DSMZ (Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig Germany) and were cultured with RPMI1640 and DMEM supplemented with 10% fetal bovine serum, penicillin (107 U/L) and streptomycin (10 mg/L) (Biochrom AG, Berlin, Germany) as previously described.[9,10]

Maximum AK release was obtained by incubating target and effector cells with Total-Lysis Reagent™ (Lonza, Cologne, Germany), and baseline AK release was obtained by incubating cells with medium alone. Baseline release from T cells and tumor cells were < 10% of maximum release in all experiments, and baseline value was subtracted from each value.

The activity of AK was determined by detection of auto-luminescence using a luciferase assay (ToxiLight Kit, Lonza, Cologne, Germany). Supernatant (20 μL) was incubated with AK-detection reagent (Lonza, Cologne, Germany) for 5 min at room temperature. Bioluminescence was measured by a luminometer (BD Monolight 3096 Microplate Luminometer, BD Biosciences, Heidelberg, Germany) and expressed as relative luminescence units (RLU).

Statistical analysis

The stimulation index (SI) of CD8+ T cells was determined by the ratio of IFNγ+CD8+ vs. IFNγ-CD8+ T cells stimulated with tumor tissue lysate vs. stimulation with normal liver tissue, respectively, and calculated with Excel® 2003 software (Microsoft, Seattle, USA). Results were analyzed statistically by the SPSS® software package (Version 14, SPSS GmbH Software, Munich, Germany). SI values were divided into two groups according to the histological origin of the tumors. SI values were calculated numerically and are presented as columns. RLU were expressed numerically and are shown as columns. The significance of the enhanced SI after RFA treatment and augmented cytotoxic activity was tested with Fisher’s test for dependent samples. P < 0.05 were considered significant.

Results

Of 3 patients suffering from CRC, 2 had a recurrence-free survival and 1 developed secondary metastases. One patient with HCC developed a local recurrence, and the other one had a disease-free survival after 12 months [Table 1].

Table 1

Summary of general data with entity of cirrhosis and entity of primary in case of metastases

PatientEntityStage of disease 12 months after RFA
Primary
CRC1 Colon descendingDisease free
CRC2 RectumMetastases to lung
CRC3 RectumDisease free
Cirrhosis
HCC1 AlcoholDisease free
HCC2 AlcoholLocal recurrence

All patients had a significant activation of tumor-specific T cells (SI baseline = 2.02, SD ± 0.2; SI CRC at 12 months = 12.3, SD ± 0.14; SI HCC at 12 months = 11.8, SD ± 0.23) (P < 0.05) [Figure 1].

Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma

Figure 1. Stimulation index of CD8+ T cells was determined by the ratio of IFNγ+CD8+ vs. IFNγ-CD8+ T cells after stimulation with autologous tumor antigens. Shown are mean ± SD. IFNγ: interferon gamma; CRC: colorectal cancer; HCC: hepatocellular carcinoma; RFA: radiofrequency ablation

Disease-free patients showed a significantly risen cytolytic activity of PBMC against matched tumor cells even 12 months after treatment (P < 0.05). The cytolytic activity after 12 months was comparable to baseline data (RLU before RFA = 10.4, SD ± 1.3; RLU 4 weeks = 354.7, SD ± 42.1; RLU CRC 12 months = 298.4, SD ± 23.1; RLU HCC 12 months = 317.4). In contrast, patients with recurrence of malignancy showed a significantly decreased cytolytic activity (RLU for CRC at 12 months = 76.2; RLU for HCC at 12 months = 102.5) (P < 0.05) [Figure 2].

Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma

Figure 2. Cytolytic activity of T cells measured by adenylate kinase (AK) release assay. AK release was quantifi ed by luminescence and expressed as RLU. A significant difference between patients with recurrence of malignancy and patients with disease-free survival is shown after 12 months (P < 0.05) only. Shown are mean ± SD. RFA: radiofrequency ablation; RLU: relative luminescence units

Discussion

In the last few years, local ablative therapies such as laser induced or radiofrequency induced thermal ablation have become more attractive as therapeutic options for patients with unresectable solid tumors. The results of local ablative therapies have shown encouraging survival rates similar to R0 resection of tumors.[1,2,11] In addition, T cell vaccination or vaccination with dendritic cells is regarded as a promising strategy for the treatment of various tumor types such as malignant melanomas,[12-20] and high expectations have been placed on cytokine-modulated immunotherapy for liver tumors.

It is well-known that RFA can induce an unspecific immune stimulation. Thus, thermal coagulation causes an inflammatory reaction with lymph-plasma-cellular infiltration that can be visualized as a hypervascular rim in contrast CT and contrast-enhanced ultrasound. This hypervascular rim can be so intense as to impede proper assessment of treatment success.[5] We recently demonstrated a specific T cell response, after RFA application, toward the orthotopic VX 2-tumor implantation in rabbit livers.[6] In the clinical setting, it was also shown that a tumor-specific immune response could be detected in patients after RFA. Our preliminary study showed a significant appearance of tumor-specific CD4+ and CD8+ T cells in peripheral blood up to 8 weeks after RFA. The cytolytic activity of isolated PBMC was also significantly elevated.[8]

In this study, we investigated the presence of tumor-specific T cells and the cytolytic activity 12 months after RFA treatment. As these tissue samples were obtained by fine-needle biopsy prior to RFA, we decided to concentrate on the measurement of CD8+ cells. These cells are reported to be the most promising cell population for anti-tumor therapies. All patients still had significantly increased levels of CD8+ cells containing cytotoxic T cells, NK cell and NK T cells as well. Whether these observed cells are resting T cells or any other activated form of T cell remains unclear.

We observed no differences between patients with recurrence of malignancy or with disease-free survival. Interestingly, cytolytic activity was significantly lower in the two patients with tumor recurrence compared. Perhaps, recurrence of malignancy is caused by an ineffective cytolytic/cytotoxic activity of CD8+ cells but needs to be confirmed with further investigations.

Prior data showed an enhanced tumor growth if RFA or resection of liver metastases of CRC were not properly performed with remaining micrometastases due to hypoxia and growth factors induced by the process of wound healing.[21,22] This might indicate that the immune response is not strong enough to control tumor growth or that the immune response is not tumor specific or not effective by the lack of tumor-specific cytolytic cells.

Furthermore, the potential role of a tumor-specific immune response as a target for supportive adjuvant therapy after RFA or resection of metastases has not yet been addressed. Therefore, larger groups with a follow-up for more than 6-12 months are needed to clarify whether lower cytolytic activity levels are correlated with recurrence of malignancy after local ablative therapies. This would suggest the need for an effective combination of drugs with RFA to achieve a long lasting strong cytolytic activity of the Th2 immune response.

Further studies are needed to investigate the potential of the cytotoxic activity of CD8+ cells as a prognostic factor. The cytotoxic activity may also play an important role in tumor recurrence. In addition, the possible correlation between an active immune response and disease free survival needs to be clarified. The combination of toll-like receptor agonists with RFA has the potential to improve the outcome of patients with solid tumors in the liver.[23]

Financial support and sponsorship

This project was supported by the Hans-Loewel foundation Bamberg, Germany.

Conflicts of interest

There are no conflicts of interest.

REFERENCES

1. Solbiati L, Ierace T, Tonolini M, Osti V, Cova L. Radiofrequency thermal ablation of hepatic metastases. Eur J Ultrasound 2001;13:149-58.

2. Solbiati L, Livraghi T, Goldberg SN, Ierace T, Meloni F, Dellanoce M, Cova L, Halpern EF, Gazelle GS. Percutaneous radio-frequency ablation of hepatic metastases from colorectal cancer: long-term results in 117 patients. Radiology 2001;221:159-66.

3. Dewhirst MW, Landon CD, Hofmann CL, Stauffer PR. Novel approaches to treatment of hepatocellular carcinoma and hepatic metastases using thermal ablation and thermosensitive liposomes. Surg Oncol Clin N Am 2013;22:545-61.

4. Sofocleous CT, Sideras P, Petre EN. "How we do it" - a practical approach to hepatic metastases ablation techniques. Tech Vasc Interv Radiol 2013;16:219-29.

5. Hansler J, Neureiter D, Strobel D, Muller W, Mutter D, Bernatik T, Hahn EG, Becker D. Cellular and vascular reactions in the liver to radio-frequency thermo-ablation with wet needle applicators. Study on juvenile domestic pigs. Eur Surg Res 2002;34:357-63.

6. Wissniowski TT, Hänsler J, Neureiter D, Frieser M, Schaber S, Esslinger B, Voll R, Strobel D, Hahn EG, Schuppan D. Activation of tumor-specific T lymphocytes by radio-frequency ablation of the VX2 hepatoma in rabbits. Cancer Res 2003;63:6496-500.

7. Behm B, Di Fazio P, Michl P, Neureiter D, Kemmerling R, Hahn EG, Strobel D, Gress T, Schuppan D, Wissniowski TT. Additive antitumour response to the rabbit VX2 hepatoma by combined radio frequency ablation and toll like receptor 9 stimulation. Gut 2014; doi: 10.1136/gutjnl-2014-308286.

8. Hansler J, Wissniowski TT, Schuppan D, Witte A, Bernatik T, Hahn EG, Strobel D. Activation and dramatically increased cytolytic activity of tumor specific T lymphocytes after radio-frequency ablation in patients with hepatocellular carcinoma and colorectal liver metastases. World J Gastroenterol 2006;12:3716-21.

9. Ocker M, Alajati A, Ganslmayer M, Zopf S, Luders M, Neureiter D, Hahn EG, Schuppan D, Herold C. The histone-deacetylase inhibitor SAHA potentiates proapoptotic effects of 5-fluorouracil and irinotecan in hepatoma cells. J Cancer Res Clin Oncol 2005;131:385-94.

10. Di Fazio P, Schneider-Stock R, Neureiter D, Okamoto K, Wissniowski T, Gahr S, Quint K, Meissnitzer M, Alinger B, Montalbano R, Sass G, Hohenstein B, Hahn EG, Ocker M. The pan-deacetylase inhibitor panobinostat inhibits growth of hepatocellular carcinoma models by alternative pathways of apoptosis. Cell Oncol 2010;32:285-300.

11. Livraghi T, Goldberg SN, Solbiati L, Meloni F, Ierace T, Gazelle GS. Percutaneous radio-frequency ablation of liver metastases from breast cancer: initial experience in 24 patients. Radiology 2001;220:145-9.

12. Berger TG, Haendle I, Schrama D, Luftl M, Bauer N, Pedersen LO, Schuler-Thurner B, Hohenberger W, Straten Pt P, Schuler G, Becker JC. Circulation and homing of melanoma-reactive T cells to both cutaneous and visceral metastases after vaccination with monocyte-derived dendritic cells. Int J Cancer 2004;111:229-37.

13. Erdmann M, Dorrie J, Schaft N, Strasser E, Hendelmeier M, Kampgen E, Schuler G, Schuler-Thurner B. Effective clinical-scale production of dendritic cell vaccines by monocyte elutriation directly in medium, subsequent culture in bags and final antigen loading using peptides or RNA transfection. J Immunother 2007;30:663-74.

14. Schaft N, Dorrie J, Muller I, Beck V, Baumann S, Schunder T, Kampgen E, Schuler G. A new way to generate cytolytic tumor-specific T cells: electroporation of RNA coding for a T cell receptor into T lymphocytes. Cancer Immunol Immunother 2006;55:1132-41.

15. Schuler G, Thurner B, Romani N. Dendritic cells: from ignored cells to major players in T-cell-mediated immunity. Int Arch Allergy Immunol 1997;112:317-22.

16. Schuler-Thurner B, Dieckmann D, Keikavoussi P, Bender A, Maczek C, Jonuleit H, Roder C, Haendle I, Leisgang W, Dunbar R, Cerundolo V, von Den Driesch P, Knop J, Brocker EB, Enk A, Kampgen E, Schuler G. Mage-3 and influenza-matrix peptide-specific cytotoxic T cells are inducible in terminal stage HLA-A2.1+melanoma patients by mature monocyte-derived dendritic cells. J Immunol 2000;165:3492-6.

17. Steinbrink K, Jonuleit H, Muller G, Schuler G, Knop J, Enk AH. Interleukin-10-treated human dendritic cells induce a melanoma-antigen-specific anergy in CD8(+) T cells resulting in a failure to lyse tumor cells. Blood 1999;93:1634-42.

18. Thurner B, Haendle I, Roder C, Dieckmann D, Keikavoussi P, Jonuleit H, Bender A, Maczek C, Schreiner D, von den Driesch P, Brocker EB, Steinman RM, Enk A, Kampgen E, Schuler G. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med 1999;190:1669-78.

19. Jarver P, Zaghloul EM, Arzumanov AA, Saleh AF, McClorey G, Hammond SM, Hallbrink M, Langel U, Smith CI, Wood MJ, Gait MJ, Andaloussi SE. Peptide nanoparticle delivery of charge-neutral splice-switching morpholino oligonucleotides. Nucleic Acid Ther 2015;25:65-77.

20. Khairuddin N, Gantier MP, Blake SJ, Wu SY, Behlke MA, Williams BR, McMillan NA. siRNA-induced immunostimulation through TLR7 promotes antitumoral activity against HPV-driven tumors in vivo. Immunol Cell Biol 2012;90:187-96.

21. van der Bilt JD, Soeters ME, Duyverman AM, Nijkamp MW, Witteveen PO, van Diest PJ, Kranenburg O, Borel Rinkes IH. Perinecrotic hypoxia contributes to ischemia/reperfusion-accelerated outgrowth of colorectal micrometastases. Am J Pathol 2007;170:1379-88.

22. Nijkamp MW, van der Bilt JD, de Bruijn MT, Molenaar IQ, Voest EE, van Diest PJ, Kranenburg O, Borel Rinkes IH. Accelerated perinecrotic outgrowth of colorectal liver metastases following radiofrequency ablation is a hypoxia-driven phenomenon. Ann Surg 2009;249:814-23.

23. Prud'homme GJ. DNA vaccination against tumors. J Gene Med 2005;7:3-17.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Wissniowski TT, Gress T, Fazio PD. Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma. Hepatoma Res 2015;1:92-6. http://dx.doi.org/10.4103/2394-5079.158024

AMA Style

Wissniowski TT, Gress T, Fazio PD. Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma. Hepatoma Research. 2015; 1: 92-6. http://dx.doi.org/10.4103/2394-5079.158024

Chicago/Turabian Style

Wissniowski, Thaddeus Till, Thomas Gress, Pietro Di Fazio. 2015. "Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma" Hepatoma Research. 1: 92-6. http://dx.doi.org/10.4103/2394-5079.158024

ACS Style

Wissniowski, TT.; Gress T.; Fazio PD. Long-term immune-modulatory side effects of radiofrequency ablation in patients with liver metastases and hepatocellular carcinoma. Hepatoma. Res. 2015, 1, 92-6. http://dx.doi.org/10.4103/2394-5079.158024

About This Article

This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License (http://creativecommons.org/licenses/by-nc-sa/3.0/), which allows others to remix, tweak and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

Data & Comments

Data

Views
8972
Downloads
917
Citations
0
Comments
0
28

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 4 clicks
Like This Article 28 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Hepatoma Research
ISSN 2454-2520 (Online) 2394-5079 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/