We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Preparation and evaluation of etoposide-loaded lipid-based nanosuspensions for high-dose treatment of lymphoma

    Xiaolan Yin

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Leiqiang Han

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Shengjun Mu

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    ,
    Weiwei Mu

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    ,
    Shuang Liang

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    ,
    Tianqi Wang

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    ,
    Yongjun Liu

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    &
    Na Zhang

    *Author for correspondence: Tel.: +86 0531 8838 2015; Fax: +86 0531 8838 2548;

    E-mail Address: zhangnancy9@sdu.edu.cn

    Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Culture Road, Ji’nan, Shandong Province 250012, PR China

    Published Online:https://doi.org/10.2217/nnm-2018-0502

    Aim: High-dose administration of etoposide (VP16) was limited by its poor aqueous solubility and severe systemic toxicity on lymphoma therapy. Herein, a novel VP16-loaded lipid-based nanosuspensions (VP16-LNS) was developed for improving drug solubility, enhancing antitumor effect and reducing systemic toxicity. Materials & methods: VP16-LNS with soya lecithin and D-α-tocopheryl PEG 1000 succinate (TPGS) as stabilizers were prepared by nanoprecipitation method. Results: VP16-LNS exhibited uniform spherical morphology, small particle size and favorable colloidal stability. The concentration of VP16 in VP16-LNS was high enough (1017.67 μg/ml) for high-dose therapy on lymphoma. Moreover, VP16-LNS displayed long blood circulation time, selective intratumoral accumulation, remarkable antitumor effect and upregulated safety. Conclusion: VP16-LNS would be an efficient nanoformulation for clinical intravenous application against lymphoma.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Teras LR, DeSantis CE, Cerhan JR, Morton LM, Jemal A, Flowers CR. 2016 US lymphoid malignancy statistics by World Health Organization subtypes. CA Cancer J. Clin. 66, 443–459 (2016).
    • 2. Ansell SM. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 91(4), 434–442 (2016).
    • 3. Stephenson R, Singh A. Drug discovery and therapeutic delivery for the treatment of B and T cell tumors. Adv. Drug Deliv. Rev. 114, 285–300 (2017).
    • 4. Zhou P, Liu P, Zhou SY et al. Ifosfamide, cisplatin or carboplatin, and etoposide (ICE)-based chemotherapy for mobilization of autologous peripheral blood stem cells in patients with lymphomas. Chin. Med. J. 128(18), 2498–2504 (2015).
    • 5. Reddy LH, Adhikari JS, Dwarakanath BSR, Sharma RK, Murthy RR. Tumoricidal effects of etoposide incorporated into solid lipid nanoparticles after intraperitoneal administration in Dalton's lymphoma bearing mice. AAPS J. 8(2), E254–E262 (2006).
    • 6. Wang HY, Sun GD, Zhang ZG, Ou Y. Transcription activator, hyaluronic acid and tocopheryl succinate multi-functionalized novel lipid carriers encapsulating etoposide for lymphoma therapy. Biomed. Pharmacother. 91, 241–250 (2017).
    • 7. Hande KR. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 34(10), 1514–1521 (1998). •• Critical developments in etoposide’s mechanism of action, pharmacology and administration schedule are summarized.
    • 8. Mahindra A, Bolwell BJ, Rybicki L et al. Etoposide plus G-CSF priming compared with G-CSF alone in patients with lymphoma improves mobilization without an increased risk of secondary myelodysplasia and leukemia. Bone Marrow Transpl. 47(2), 231–235 (2012).
    • 9. Green DJ, Bensinger WI, Holmberg LA et al. Bendamustine, etoposide and dexamethasone to mobilize peripheral blood hematopoietic stem cells for autologous transplantation in patients with multiple myeloma. Bone Marrow Transpl. 51(10), 1330–1336 (2016).
    • 10. Al-Ali AAA, Quach JRC, Bundgaard C, Steffansen B, Holm R, Nielsen CU. Polysorbate 20 alters the oral bioavailability of etoposide in wild type and mdr1a deficient Sprague-Dawley rats. Int. J. Pharm. 543(1–2), 352–360 (2018).
    • 11. Pooja D, Kulhari H, Tunki L et al. Nanomedicines for targeted delivery of etoposide to non-small cell lung cancer using transferrin functionalized nanoparticles. RSC Adv. 5(61), 49122–49131 (2015).
    • 12. Ozkan HA, Bal C, Gulbas Z. Chemomobilization with high-dose etoposide and G-CSF results in effective and safe stem cell collection in heavily pretreated lymphoma patients: report from a single institution study and review. Eur. J. Hematol. 92(5), 390–397 (2014).
    • 13. Agrahari V, Agrahari V. Facilitating the translation of nanomedicines to a clinical product: challenges and opportunities. Drug Discov. Today 23(5), 974–991 (2018).
    • 14. Youn YS, Bae YH. Perspectives on the past, present, and future of cancer nanomedicine. Adv. Drug Deliv. Rev. 130, 3–11 (2018).
    • 15. Duan R, Li CY, Wang F, Yangi JC. Polymer–lipid hybrid nanoparticles-based paclitaxel and etoposide combinations for the synergistic anticancer efficacy in osteosarcoma. Colloid Surf. B. 159, 880–887 (2017).
    • 16. Wojnilowicz M, Tortora M, Bobay BG et al. A combined approach for predicting the cytotoxic effect of drug-nanoaggregates. J. Mater. Chem. B 4(40), 6516–6523 (2016).
    • 17. Qu D, Wang L, Liu M et al. Oral nanomedicine based on multicomponent microemulsions for drug-resistant breast cancer treatment. Biomacromolecules 18(4), 1268–1280 (2017).
    • 18. Wei X, Senanayake TH, Warren G, Vinogradov SV. Hyaluronic acid-based nanogel–drug conjugates with enhanced anticancer activity designed for the targeting of CD44-positive and drug-resistant tumors. Bioconjug. Chem. 24(4), 658–668 (2013).
    • 19. Mishra DK, Shandilya R, Mishra PK. Lipid based nanocarriers: a translational perspective. Nanomedicine 14(7), 2023–2050 (2018). • The latest advances and updates in lipidic nanocarriers, their formulation strategies, challenging aspects, stability profile, clinical applications alongside commercially available products and products under clinical trials.
    • 20. Yang L, Hong J, Di J et al. 10-hydroxycamptothecin (HCPT) nanosuspensions stabilized by mPEG1000-HCPT conjugate: high stabilizing efficiency and improved antitumor efficacy. Int. J. Nanomed. 12, 3681–3695 (2017).
    • 21. Ahire E, Thakkar S, Darshanwad M, Misra M. Parenteral nanosuspensions: a brief review from solubility enhancement to more novel and specific applications. Acta Pharm. Sin. B 8(5), 733–755 (2018). •• Focuses on parenteral nanosuspensions, covering varied aspects, especially stabilizers used, scalability challenges, issues of physical and chemical stability, solidification techniques to combat stability problems, and in vivo fate.
    • 22. Wang L, Liu Z, Liu D, Liu C, Juan Z, Zhang N. Docetaxel-loaded-lipid-based-nanosuspensions (DTX-LNS): preparation, pharmacokinetics, tissue distribution and antitumor activity. Int. J. Pharm. 413(1–2), 194–201 (2011).
    • 23. Wang L, Li M, Zhang N. Folate-targeted docetaxel-lipid-based-nanosuspensions for active-targeted cancer therapy. Int. J. Nanomed. 7, 3281–3294 (2012).
    • 24. Yang SM, Zhang B, Gong XW, Wang TQ, Liu YJ, Zhang N. In vivo biodistribution, biocompatibility, and efficacy of sorafenib-loaded lipid-based nanosuspensions evaluated experimentally in cancer. Int. J. Nanomed. 11, 2329–2343 (2016).
    • 25. Zhang J, Li M, Liu Z, Wang L, Liu Y, Zhang N. Preclinical studies of N3-O-toluyl-fluorouracil-loaded lipid-based nanosuspensions in H22-bearing mice. Int. J. Nanomed. 9, 2741–2751 (2014).
    • 26. Wang T, Feng L, Yang S, Liu Y, Zhang N. Ceramide lipid-based nanosuspension for enhanced delivery of docetaxel with synergistic antitumor efficiency. Drug Deliv. 24(1), 800–810 (2017).
    • 27. Zhang J, Wang TQ, Mu SJ, Olerile LD, Yu XY, Zhang N. Biomacromolecule/lipid hybrid nanoparticles for controlled delivery of sorafenib in targeting hepatocellular carcinoma therapy. Nanomedicine 12(8), 911–925 (2017).
    • 28. Zhang ZP, Tan SW, Feng SS. Vitamin E TPGS as a molecular biomaterial for drug delivery. Biomaterials 33(19), 4889–4906 (2012). • Reviews the physicochemical and pharmaceutical properties of vitamin E tocopheryl PEG 1000 succinate (TPGS) and its wide applications in composition of the various nanocarriers for drug delivery.
    • 29. Duhem N, Danhier F, Preat V. Vitamin E-based nanomedicines for anti-cancer drug delivery. J. Control. Rel. 182, 33–44 (2014).
    • 30. Wang L, Liu Y, Zhao J et al. In vitro and in vivo evaluation of targeting tumor with folate-based amphiphilic multifunctional stabilizer for resveratrol nanosuspensions. Colloids Surf. B Biointerfaces 160, 462–472 (2017).
    • 31. Yin X, Xiao Y, Han L et al. Ceramide-fabricated co-loaded liposomes for the synergistic treatment of hepatocellular carcinoma. AAPS PharmSciTech 19(5), 2133–2143 (2018).
    • 32. Zhang B, Wang TQ, Yang SM et al. Development and evaluation of oxaliplatin and irinotecan co-loaded liposomes for enhanced colorectal cancer therapy. J. Control. Rel. 238, 10–21 (2016).
    • 33. Zhai Q, Chen Y, Xu J et al. Lymphoma immunochemotherapy: targeted delivery of doxorubicin via a dual functional nanocarrier. Mol. Pharm. 14(11), 3888–3895 (2017).
    • 34. Fang T, Duarte JN, Ling JJ, Li ZY, Guzman JS, Ploegh HL. Structurally defined aMHC-II nanobody–drug conjugates: a therapeutic and imaging system for b-Cell lymphoma. Angew. Chem. Int. Ed. Engl. 55(7), 2416–2420 (2016).
    • 35. Papiez MA, Krzysciak W, Szade K et al. Curcumin enhances the cytogenotoxic effect of etoposide in leukemia cells through induction of reactive oxygen species. Drug Des. Dev. Ther. 10, 557–570 (2016).
    • 36. Yordanov G, Skrobanska R, Evangelatov A. Colloidal formulations of etoposide based on poly(butyl cyanoacrylate) nanoparticles: preparation, physicochemical properties and cytotoxicity. Colloids Surf. B Biointerfaces 101, 215–222 (2013).
    • 37. Hao JF, Gao Y, Zhao J et al. Preparation and optimization of resveratrol nanosuspensions by antisolvent precipitation using Box–Behnken design. AAPS PharmSciTech 16(1), 118–128 (2015).
    • 38. Deshantri AK, Varela Moreira A, Ecker V et al. Nanomedicines for the treatment of hematological malignancies. J. Control. Rel. 287, 194–215 (2018). • Describes currently available therapies as well as limitations and potential advantages of nanomedicine formulations for treatment of hematological malignancies.
    • 39. Joel SP, Shah R, Slevin ML. Etoposide dosage and pharmacodynamics. Cancer Chemother. Pharmacol. 34, S69–S75 (1994).
    • 40. Guo Y, Luo J, Tan S, Otieno BO, Zhang Z. The applications of vitamin E TPGS in drug delivery. Eur. J. Pharm. Sci. 49(2), 175–186 (2013).
    • 41. Dai Y, Meng Q, Mu W, Zhang T. Recent advances in the applications and biotechnological production of mannitol. J. Funct. Foods 36, 404–409 (2017).
    • 42. Thorat AA, Dalvi SV. Liquid antisolvent precipitation and stabilization of nanoparticles of poorly water soluble drugs in aqueous suspensions: recent developments and future perspective. Chem. Eng. J. 181, 1–34 (2012).
    • 43. Jamrógiewicz M, Pieńkowska K. Recent breakthroughs in the stability testing of pharmaceutical compounds. Trends Anal. Chem. 111, 118–127 (2019).
    • 44. Wang Y, Zheng Y, Zhang L, Wang Q, Zhang D. Stability of nanosuspensions in drug delivery. J. Control. Rel. 172(3), 1126–1141 (2013).
    • 45. Rewatkar PV, Parton RG, Parekh HS, Parat MO. Are caveolae a cellular entry route for non-viral therapeutic delivery systems? Adv. Drug Deliv. Rev. 91, 92–108 (2015).
    • 46. Tao Z, Dang X, Huang X et al. Early tumor detection afforded by in vivo imaging of near-infrared II fluorescence. Biomaterials 134, 202–215 (2017).
    • 47. Etrych T, Lucas H, Janouskova O, Chytil P, Mueller T, Mader K. Fluorescence optical imaging in anticancer drug delivery. J. Control. Rel. 226, 168–181 (2016).
    • 48. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliv. Rev. 91, 3–6 (2015).
    • 49. Kolate A, Baradia D, Patil S, Vhora I, Kore G, Misra A. PEG – a versatile conjugating ligand for drugs and drug delivery systems. J. Control. Rel. 192, 67–81 (2014).
    • 50. Choudhury H, Gorain B, Pandey M et al. Recent advances in TPGS-based nanoparticles of docetaxel for improved chemotherapy. Int. J. Pharm. 529(1-2), 506–522 (2017).
    • 51. Mishra P, Nayak B, Dey RK. PEGylation in anti-cancer therapy: an overview. Asian J. Pharm. Sci. 11(3), 337–348 (2016).
    • 52. Walkey CD, Chan WC. Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. Chem. Soc. Rev. 41(7), 2780–2799 (2012).
    • 53. Bascuas T, Moreno M, Monaco A et al. A novel non-Hodgkin lymphoma murine model closer to the standard clinical scenario. J. Transl. Med. 14(1), 323–335 (2016).
    • 54. Mu S, Liu Y, Wang T et al. Unsaturated nitrogen-rich polymer poly(l-histidine) gated reversibly switchable mesoporous silica nanoparticles using “graft to” strategy for drug controlled release. Acta Biomater. 63, 150–162 (2017).
    • 55. Hare JI, Lammers T, Ashford MB, Puri S, Storm G, Barry ST. Challenges and strategies in anti-cancer nanomedicine development: an industry perspective. Adv. Drug Deliv. Rev. 108, 25–38 (2017). •• The challenges and strategies of anticancer nanomedicine for the successful translation from preclinical proof of concept to demonstration of therapeutic value in the clinic.
    • 56. Tinkle S, McNeil SE, Muhlebach S et al. Nanomedicines: addressing the scientific and regulatory gap. Ann. NY Acad. Sci. 1313, 35–56 (2014).
    • 57. Muhlebach S. Regulatory challenges of nanomedicines and their follow-on versions: a generic or similar approach? Adv. Drug Deliv. Rev. 131, 122–131 (2018).