Skip to main content

Advertisement

Log in

Conventional versus drug-eluting embolic transarterial chemoembolization with doxorubicin: comparative drug delivery, pharmacokinetics, and treatment response in a rabbit VX2 tumor model

  • Original Article
  • Published:
Drug Delivery and Translational Research Aims and scope Submit manuscript

Abstract

The purpose of this study was to compare intra-tumoral drug delivery, pharmacokinetics, and treatment response after doxorubicin (DOX) conventional (c-) versus drug-eluting embolic (DEE-) transarterial chemoembolization (TACE) in a rabbit VX2 liver tumor model. Twenty-four rabbits with solitary liver tumors underwent c-TACE (n = 12) (1:2 water-in-oil emulsion, 0.6 mL volume, 2 mg DOX) or DEE-TACE (n = 12) (130,000 70–150 µm 2 mg DOX-loaded microspheres). Systemic, intra-tumoral, and liver DOX levels were measured using mass spectrometry up to 7-day post-procedure. Intra-tumoral DOX distribution was quantified using fluorescence imaging. Percent tumor necrosis was quantified by a pathologist blinded to treatment group. Lobar TACE was successfully performed in all cases. Peak concentration (CMAX, µg/mL) for plasma, tumor tissue, and liver were 0.666, 4.232, and 0.270 for c-TACE versus 0.103, 8.988, and 0.610 for DEE-TACE. Area under the concentration versus time curve (AUC, µg/mL ∗ min) for plasma, tumor tissue, and liver were 18.3, 27,078.8, and 1339.1 for c-TACE versus 16.4, 26,204.8, and 1969.6 for DEE-TACE. A single dose of intra-tumoral DOX maintained cytotoxic levels through 7-day post-procedure for both TACE varieties, with a half-life of 1.8 (c-TACE) and 0.8 (DEE-TACE) days. Tumor-to-normal liver DOX ratio was high (c-TACE, 20.2; DEE-TACE, 13.3). c-TACE achieved significantly higher DOX coverage of tumor vs. DEE-TACE (10.8% vs. 2.3%; P = 0.003). Percent tumor necrosis was similar (39% vs. 37%; P = 0.806). In conclusion, in a rabbit VX2 liver tumor model, both c-TACE and DEE-TACE achieved tumoricidal intra-tumoral DOX levels and high tumor-to-normal liver drug ratios, though c-TACE resulted in significantly greater tumor coverage.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data Availability

Will be considered upon request.

References

  1. Malagari K, Pomoni M, Kelekis A, Pomoni A, Dourakis S, Spyridopoulos T, et al. Prospective randomized comparison of chemoembolization with doxorubicin-eluting beads and bland embolization with BeadBlock for hepatocellular carcinoma. Cardiovasc Intervent Radiol. 2010;33(3):541–51.

    Article  Google Scholar 

  2. Shi M, Lu LG, Fang WQ, Guo RP, Chen MS, Li Y, et al. Roles played by chemolipiodolization and embolization in chemoembolization for hepatocellular carcinoma: single-blind, randomized trial. J Natl Cancer Inst. 2013;105(1):59–68.

    Article  CAS  Google Scholar 

  3. Johnson PJ, Kalayci C, Dobbs N, Raby N, Metivier EM, Summers L, et al. Pharmacokinetics and toxicity of intraarterial adriamycin for hepatocellular carcinoma: effect of coadministration of lipiodol. J Hepatol. 1991;13(1):120–7.

    Article  CAS  Google Scholar 

  4. Raoul JL, Heresbach D, Bretagne JF, Ferrer DB, Duvauferrier R, Bourguet P, et al. Chemoembolization of hepatocellular carcinomas. A study of the biodistribution and pharmacokinetics of doxorubicin. Cancer. 1992;70(3):585–90.

  5. Namur J, Wassef M, Millot JM, Lewis AL, Manfait M, Laurent A. Drug-eluting beads for liver embolization: concentration of doxorubicin in tissue and in beads in a pig model. J Vasc Interv Radiol. 2010;21(2):259–67.

    Article  Google Scholar 

  6. Dreher MR, Sharma KV, Woods DL, Reddy G, Tang Y, Pritchard WF, et al. Radiopaque drug-eluting beads for transcatheter embolotherapy: experimental study of drug penetration and coverage in swine. J Vasc Interv Radiol. 2012;23(2):257–64 e4.

  7. Zhang SHC, Li Z, Yang Y, Bao T, Chen H, Zou Y, Song L. Comparison of pharmacokinetics and drug release in tissues after transarterial chemoembolization with doxorubicin using diverse lipiodol emulsions and CalliSpheres Beads in rabbit livers. Drug Deliv. 2017;24(1):101–7.

    Google Scholar 

  8. Gupta S, Wright KC, Ensor J, Van Pelt CS, Dixon KA, Kundra V. Hepatic arterial embolization with doxorubicin-loaded superabsorbent polymer microspheres in a rabbit liver tumor model. Cardiovasc Intervent Radiol. 2011;34(5):1021–30.

    Article  Google Scholar 

  9. Choi JWCH, Park JH, Baek SY, Chung JW, Kim DD, Kim HC. Comparison of drug release and pharmacokinetics after transarterial chemoembolization using diverse lipiodol emulsions and drug-eluting beads. PLoS One. 2014;9(12):e115898.

    Article  Google Scholar 

  10. Gaba RC. Chemoembolization practice patterns and technical methods among interventional radiologists: results of an online survey. AJR Am J Roentgenol. 2012;198(3):692–9.

    Article  Google Scholar 

  11. Varela M, Real MI, Burrel M, Forner A, Sala M, Brunet M, et al. Chemoembolization of hepatocellular carcinoma with drug eluting beads: efficacy and doxorubicin pharmacokinetics. J Hepatol. 2007;46(3):474–81.

    Article  CAS  Google Scholar 

  12. Rous P, Beard JW. The Progression to Carcinoma of Virus-Induced Rabbit Papillomas (Shope). J Exp Med. 1935;62(4):523–48.

    Article  CAS  Google Scholar 

  13. Kidd JG, Rous P. A transplantable rabbit carcinoma originating in a virus-induced papilloma and containing the virus in masked or altered form. J Exp Med. 1940;71(6):813–38.

    Article  CAS  Google Scholar 

  14. Virmani S, Harris KR, Szolc-Kowalska B, Paunesku T, Woloschak GE, Lee FT, et al. Comparison of two different methods for inoculating VX2 tumors in rabbit livers and hind limbs. J Vasc Interv Radiol. 2008;19(6):931–6.

    Article  Google Scholar 

  15. Ko YH, Pedersen PL, Geschwind JF. Glucose catabolism in the rabbit VX2 tumor model for liver cancer: characterization and targeting hexokinase. Cancer Lett. 2001;173(1):83–91.

    Article  CAS  Google Scholar 

  16. Hamuro M, Nakamura K, Sakai Y, Nakata M, Ichikawa H, Fukumori Y, et al. New oily agents for targeting chemoembolization for hepatocellular carcinoma. Cardiovasc Intervent Radiol. 1999;22(2):130–4.

    Article  CAS  Google Scholar 

  17. Ramsey DE, Kernagis LY, Soulen MC, Geschwind JF. Chemoembolization of hepatocellular carcinoma. J Vasc Interv Radiol. 2002;13(9 Pt 2):S211–21.

    Article  Google Scholar 

  18. Arifin WN, Zahiruddin WM. Sample size calculation in animal studies using resource equation approach. Malays J Med Sci. 2017;24(5):101–5.

    PubMed  PubMed Central  Google Scholar 

  19. Easty DM, Easty GC. Establishment of an in vitro cell line from the rabbit VX2 carcinoma. Virchows Arch B Cell Pathol Incl Mol Pathol. 1982;39(3):333–7.

    Article  CAS  Google Scholar 

  20. Yoneda T, Kitamura M, Ogawa T, Aya S, Sakuda M. Control of VX2 carcinoma cell growth in culture by calcium, calmodulin, and prostaglandins. Cancer Res. 1985;45(1):398–405.

    CAS  PubMed  Google Scholar 

  21. Namur J, Citron SJ, Sellers MT, Dupuis MH, Wassef M, Manfait M, et al. Embolization of hepatocellular carcinoma with drug-eluting beads: doxorubicin tissue concentration and distribution in patient liver explants. J Hepatol. 2011;55(6):1332–8.

    Article  CAS  Google Scholar 

  22. Gaba RC, Baumgarten S, Omene BO, van Breemen RB, Garcia KD, Larson AC, et al. Ethiodized oil uptake does not predict doxorubicin drug delivery after chemoembolization in VX2 liver tumors. J Vasc Interv Radiol. 2012;23(2):265–73.

    Article  Google Scholar 

  23. Parvinian A, Casadaban LC, Gaba RC. Development, growth, propagation, and angiographic utilization of the rabbit VX2 model of liver cancer: a pictorial primer and “how to” guide. Diagn Interv Radiol. 2014;20(4):335–40.

    Article  Google Scholar 

  24. Kan Z, Wright K, Wallace S. Ethiodized oil emulsions in hepatic microcirculation: in vivo microscopy in animal models. Acad Radiol. 1997;4(4):275–82.

    Article  CAS  Google Scholar 

  25. Casadaban L, Parvinian A, Gaba RC. Effects of doxorubicin (DOX) delivery on tumor necrosis after drug-eluting bead transarterial chemoembolization (DEB-TACE). J Vasc Interv Radiol. 2015;26(2):S18.

    Google Scholar 

  26. Doxorubicin Material Safety Data Sheet [Available from: http://datasheets.scbt.com/sc-200923.pdf.

  27. Loading of DC Beads using Doxorubicin Powder 10mg in vial 2019 [Available from: http://bead.btg-im.com/products/uk-322/dcbead-3/dcbead-loading-debdox.

  28. Lewis AL, Gonzalez MV, Lloyd AW, Hall B, Tang Y, Willis SL, et al. DC bead: in vitro characterization of a drug-delivery device for transarterial chemoembolization. J Vasc Interv Radiol. 2006;17(2 Pt 1):335–42.

    Article  Google Scholar 

  29. Bourget P, Delouis JM. Review of a technic for the estimation of area under the concentration curve in pharmacokinetic analysis. Therapie. 1993;48(1):1–5.

    CAS  PubMed  Google Scholar 

  30. Virmani S, Rhee TK, Ryu RK, Sato KT, Lewandowski RJ, Mulcahy MF, et al. Comparison of hypoxia-inducible factor-1alpha expression before and after transcatheter arterial embolization in rabbit VX2 liver tumors. J Vasc Interv Radiol. 2008;19(10):1483–9.

    Article  Google Scholar 

  31. Manautou JE, Silva VM, Hennig GE, Whiteley HE. Repeated dosing with the peroxisome proliferator clofibrate decreases the toxicity of model hepatotoxic agents in male mice. Toxicology. 1998;127(1–3):1–10.

    Article  CAS  Google Scholar 

  32. Ramos-Vara JA, Miller MA. When tissue antigens and antibodies get along: revisiting the technical aspects of immunohistochemistry–the red, brown, and blue technique. Vet Pathol. 2014;51(1):42–87.

    Article  CAS  Google Scholar 

  33. Fox SB, Gasparini G, Harris AL. Angiogenesis: pathological, prognostic, and growth-factor pathways and their link to trial design and anticancer drugs. Lancet Oncol. 2001;2(5):278–89.

    Article  CAS  Google Scholar 

  34. Gaba RC, Elkhadragy L, Boas FE, Chaki S, Chen HH, El-Kebir M, et al. Development and comprehensive characterization of porcine hepatocellular carcinoma for translational liver cancer investigation. Oncotarget. 2020;in press.

  35. Idee JM, Guiu B. Use of Lipiodol as a drug-delivery system for transcatheter arterial chemoembolization of hepatocellular carcinoma: a review. Crit Rev Oncol Hematol. 2013;88(3):530–49.

    Article  Google Scholar 

  36. Gaba RC, Schwind RM, Ballet S. Mechanism of action, pharmacokinetics, efficacy, and safety of transarterial therapies using ethiodized oil: preclinical review in liver cancer models. J Vasc Interv Radiol. 2018;29(3):413–24.

    Article  Google Scholar 

  37. Piscitelli SC, Rodvold KA, Rushing DA, Tewksbury DA. Pharmacokinetics and pharmacodynamics of doxorubicin in patients with small cell lung cancer. Clin Pharmacol Ther. 1993;53(5):555–61.

    Article  CAS  Google Scholar 

  38. de Baere T, Arai Y, Lencioni R, Geschwind JF, Rilling W, Salem R, et al. Treatment of liver tumors with lipiodol TACE: technical recommendations from experts opinion. Cardiovasc Intervent Radiol. 2016;39(3):334–43.

    Article  Google Scholar 

  39. Chen P, Yuan P, Chen B, Sun J, Shen H, Qian Y. Evaluation of drug-eluting beads versus conventional transcatheter arterial chemoembolization in patients with unresectable hepatocellular carcinoma: a systematic review and meta-analysis. Clin Res Hepatol Gastroenterol. 2017;41(1):75–85.

    Article  CAS  Google Scholar 

  40. Liu K, Min XL, Peng J, Yang K, Yang L, Zhang XM. The changes of HIF-1alpha and VEGF expression after TACE in patients with hepatocellular carcinoma. J Clin Med Res. 2016;8(4):297–302.

    Article  CAS  Google Scholar 

  41. Khabbaz RC, Huang YH, Smith AA, Garcia KD, Lokken RP, Gaba RC. Development and angiographic use of the rabbit VX2 model for liver cancer. J Vis Exp. 2019(143).

Download references

Acknowledgements

Histological services were provided by the Research Resources Center Research Histology and Tissue Imaging Core at the University of Illinois at Chicago, established with the support of the Vice Chancellor of Research.

Funding

Study funded by Guerbet USA, LLC.

Author information

Authors and Affiliations

Authors

Contributions

All authors made substantial contributions to the conception or design of the work or the acquisition, analysis, or interpretation of data for the work; AND drafting the work or revising it critically for important intellectual content; AND final approval of the version to be published; AND agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Corresponding author

Correspondence to Ron C. Gaba.

Ethics declarations

Ethical standards

The experiments comply with the current laws of the country in which they were performed.

Competing interests

Ron C. Gaba receives research support from Guerbet USA LLC, Janssen Research & Development LLC, the US Department of Defense, and the US National Institutes of Health. He serves as a consultant for Sus Clinicals, Inc.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (TIF 3.12 MB)

Supplementary file1 (TIF 10.3 MB)

Appendices

Appendix 1. Rabbit VX2 cell line development and liver tumor generation

Frozen VX2 cells in 1-mL aliquots were defrosted for 5 min at 37 °C, mixed with 1 mL of methylcellulose medium, and were injected into the left quadriceps muscle of a New Zealand White rabbit. After 2 weeks, the resulting tumor was harvested, transected, and scraped clean of necrotic debris. Several 1–2 mm3 pieces of viable tumor were saved in room temperature saline for liver implantation. The remaining tumor was used to generate additional malignant cells which were flash frozen at −80 °C or injected into the hind limb of another donor subject.

For liver tumor generation, New Zealand White rabbits underwent anesthesia induction with ketamine (30 mg/kg) and dexmedetomidine (0.1 mg/kg), followed by endotracheal intubation. Anesthesia was maintained with 1–3% isoflurane. Enrofloxacin (5 mg/kg2) was administered for infection prophylaxis. A loading dose of meloxicam (0.2 mg/kg) was provided for pain management and was continued at 1 mg/kg meloxicam for three days post-procedure.

The abdomen was shaved, sterilized, and draped. The xiphoid process was palpated, and a subxiphoid laparotomy was performed to expose the left hepatic lobe. The left hepatic lobe was gently extracted from the peritoneum. An 11-blade was used to make an incision for tumor implantation. Next, one 1–2 mm3 piece of viable tumor was placed into the liver. The liver wound was dressed with hemostatic surgical sponge (BloodSTOP iX; PRN Pharmacal, Pensacola FL). Once hemostatasis was confirmed, the liver was returned into the abdominal cavity. The muscular and fascial layers of the abdomen were closed using absorbable suture (PDS; Ethicon, Somerville NJ), and the skin was closed with absorbable suture (Vicryl; Ethicon). The tumor was allowed to grow for 2–4 weeks per established growth timelines [41].

Appendix 2. Mass spectrometry drug quantification

The ultra-high-performance liquid chromatography (UHPLC) separation was performed on the Waters Acquity™ UPLC system (Waters Corporation, Midford MA), and the analytical column used was an Acquity UPLC™ BEH C18 (2.1 × 100 mm, 1.7 μm). The separation was performed with a solvent gradient of acetonitrile (solvent A) and 5 mM ammonium acetate in dH2O (pH adjusted to 3.5 using acetic acid) (solvent B). The linear gradient was 20–80% solvent A for 1 min, held constant for 1 min, and then equilibrated back to 20% solvent A for 1 min. The flow rate of mobile phase was set at 0.4 mL/min with column temperature of 35 °C and injection volume of 5 µL.

The UHPLC system was coupled to a Waters XEVO TQD MS system (Waters Corporation) in the positive ion mode. Data was acquired by single reaction monitoring (SRM) mode. The capillary voltage was 3.8 kV. The source block and desolvation temperatures were set at 150 °C and 500 °C, respectively. The cone gas flow was 50 L/h, and the desolvation gas flow was 650 L/h. Argon was used as the collision gas. The transitions for the SRM method used in quantitation were 544.0 to 397.0 (DOX) and 548.1 to 401.1 (13C-d3-DOX).

Serum samples were analyzed in duplicate. Briefly, 250 µL of serum was added to 750 µL of ice cold acetonitrile and vortex mixed for 4 min. The mixture was centrifuged at 13,000×g for 15 min at 4 °C. The supernatant was transferred to a clean micro-centrifuge tube and dried. Samples were reconstituted in 100 µL of 50:50 acetonitrile and ultrapure water containing 5 mM ammonium acetate (pH 3.5) and internal standard (13C-d3-DOX at 100 ng/mL). An aliquot of 5 µL was then injected into the MS unit for analysis.

Tissue samples were homogenized in a phosphate buffer (0.05 M, pH 7.4) to produce a blend containing 0.2 g tissue/mL. Homogenized tissue was then extracted with four volumes of ice cold acetonitrile and centrifuged at 13,000×g for 15 min at 4 °C. The supernatant was then removed and dried. Samples were reconstituted in 300 µL of 50:50 acetonitrile and ultrapure water containing 5 mM ammonium acetate (pH 3.5) and internal standard (13C-d3-DOX at 100 ng/mL). An aliquot of 5 µL was then injected into the MS unit for analysis. Standard curves were prepared by spiking DOX standards to blank serum or homogenized blank liver tissues. The range of DOX calibration standards was 1.2–8000 ng/mL.

Appendix 3. Histologic staining methodology

Formalin-fixed VX2 tumor samples were processed on ASP300S automated tissue processor (Leica Biosystems, Wetzlar Germany) using standard overnight processing protocol (Table 5) and embedded into paraffin blocks. Tissue was sectioned at 5 µm, and adjacent sections were stained with H&E, anti-HIF-1α [H1alpha67] antibody at 1:50 dilution (Catalog #NBS100-123; Novus Biologicals, Littleton CO), anti-VEGFA[JH121] antibody at 1:50 dilution (Catalog #ab28775; Abcam, Cambridge United Kingdom), and anti-CD31 [JC70A] antibody at 1:25 dilution (Catalog #M0823, Agilent, Santa Clara CA). For H&E, tissue sections were baked, deparaffinized, and stained on Autostainer XL (Leica Biosystems) following a preset protocol. For IHC, tissue was deparaffinized and stained on BOND RX automated stainer (Leica Biosystems) using BOND Research Detection System (#DS9455, Leica). For all markers, EDTA-based (BOND ER2 solution, pH9, #AR9640) was used to retrieve the antigen. Endogenous peroxidase activity and non-specific binding were blocked by treating samples with peroxidase block (3% H2O2 in methanol) and protein block (Background Sniper #BS966; Biocare Medical, Pacheco CA). Sections were incubated with antibodies for 1 h at room temperature, and signal detection was performed using MACH2 Mouse HRP-Polymer (#MHRP520, Biocare Medical) and Betazoid DAB Chromogen (#BDB2004, Biocare Medical). For CD31 detection, BOND DAB Enhancer (AR9432, Leica Biosystems) was applied for 10 min to strengthen the signal. All sections were then counterstained with hematoxylin for 10 min and mounted with Micromount media (#3,801,730, Leica Microsystems). Secondary antibody only controls were performed to confirm the specificity of the staining. Whole slide images were acquired at × 20 magnification on an Aperio AT2 digital automated scanner (Leica Biosystems).

Table 5 Standard overnight processing protocol

Appendix 4. IHC scoring

Score

Nuclear staining (HIF-1α)

Cytoplasmic staining (VEGF)

0

0% positive cells

No staining

1

1–10% positive cells

Weak staining in < 50% cells

2

11–50% positive cells

Strong staining in < 50% cells or weak staining in > 50% cells

3

51–100% positive cells

Strong staining in > 50% cells

  1. IHC  immunohistochemistry, HIF-1α hypoxia inducible factor-1α, VEGF  vascular endothelial growth factor

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gaba, R.C., Khabbaz, R.C., Muchiri, R.N. et al. Conventional versus drug-eluting embolic transarterial chemoembolization with doxorubicin: comparative drug delivery, pharmacokinetics, and treatment response in a rabbit VX2 tumor model. Drug Deliv. and Transl. Res. 12, 1105–1117 (2022). https://doi.org/10.1007/s13346-021-00985-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13346-021-00985-8

Keywords

Navigation