Skip to main content

Advertisement

Log in

Utilization of Lipophilic Salt and Phospholipid Complex in Lipid-Based Formulations to Modulate Drug Loading and Oral Bioavailability of Pazopanib

  • Research Article
  • Published:
AAPS PharmSciTech Aims and scope Submit manuscript

Abstract

Pazopanib hydrochloride (PAZ) displays strong intermolecular interaction in its crystal lattice structure, limiting its solubility and dissolution. The development of lipid-based formulations (LbFs) resulted in reduced PAZ loading due to solid-state mediated low liposolubility. This study aims to enhance our understanding of PAZ crystallinity by synthesizing a lipophilic salt and phospholipid complex and investigating its impact on the drug loading in LbFs. The synthesized pazopanib lipophilic salt and phospholipid complex were extensively characterized. The solid form of pazopanib docusate (PAZ-DOC) and pazopanib phospholipid complex (PAZ-PLC) indicates a reduction in characteristic diffraction peaks of crystalline PAZ. The lipid formulations were prepared using synthesized PAZ-DOC and PAZ-PLC, where PAZ-DOC demonstrated six fold higher drug solubility than the commercial salt form and twice that of the PAZ-PLC due to differences in the crystallinity. Further, the impact of salt and complex formation was assessed on the aqueous drug solubilization using lipolysis and multimedia dissolution experiments. Moreover, the LbFs showed notably faster dissolution compared to the crystalline PAZ and marketed tablet. In terms of in vivo pharmacokinetics, the PAZ-DOC LbF exhibited a remarkable 11-fold increase in AUC value compared to the crystalline PAZ and a 2.5-fold increase compared to Votrient®. Similarly, PAZ-PLC LbF showed an approximately nine fold increase in drug exposure compared to the crystalline PAZ, and a 2.2-fold increase compared to Votrient®. These findings suggest that disrupting the crystallinity of drugs and incorporating them into LbF could be advantageous for enhancing drug loading and overcoming limitations related to drug absorption.

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
Fig. 8

Similar content being viewed by others

Data Availability

Data will be made available on request.

Abbreviations

AUC:

Area under the curve

BCS:

Biopharmaceutical classification system

DOC:

Docusate

DLS:

Dynamic light scattering

FTIR:

Fourier transform infrared

GI:

Gastrointestinal

GRAS:

Generally recognized as safe

HRMS:

High-resolution mass spectrometry

HTP:

High throughput

LbFs:

Lipid-based formulations

1H-NMR:

Nuclear magnetic resonance

PAZ:

Pazopanib hydrochloride

PAZ-DOC:

Pazopanib docusate

PAZ-PLC:

Pazopanib phospholipid complex

PDGFR:

Platelet-derived growth factor receptors

PLC:

Phospholipid complex

SEM:

Scanning electron microscopy

PDI:

Polydispersity index

PLM:

Polarized light microscopy

TEM:

Transmission electron microscopy

VEGFR:

Vascular endothelial growth factor receptors

References

  1. Nadaf SJ, Killedar SG, Kumbar VM, Bhagwat DA, Gurav SS. Pazopanib-laden lipid based nanovesicular delivery with augmented oral bioavailability and therapeutic efficacy against non-small cell lung cancer. Int J Pharm. 2022;628:122287.

    Article  CAS  PubMed  Google Scholar 

  2. US Food and Drug administration (FDA), 2009 [Internet]. [cited 2024 Jan 11]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/022465s000_ClinPharmR.pdf. Accessed 11 Jan 2024.

  3. Muhammad HJ, Shimada T, Fujita A, Sai Y. Sodium citrate buffer improves pazopanib solubility and absorption in gastric acid-suppressed rat model. Drug Metab Pharmacokinet [Internet]. 2024 [cited 2024 Jan 8];100995. Available from: https://www.sciencedirect.com/science/article/pii/S1347436724000016. Accessed 8 Jan 2024.

  4. Jede C, Wagner C, Kubas H, Weigandt M, Weber C, Lecomte M, et al. Improved prediction of in vivo supersaturation and precipitation of poorly soluble weakly basic drugs using a biorelevant bicarbonate buffer in a gastrointestinal transfer model. Mol Pharm. 2019;16:3938–47. https://doi.org/10.1021/acs.molpharmaceut.9b00534.

    Article  CAS  PubMed  Google Scholar 

  5. Herbrink M, Groenland SL, Huitema ADR, Jan HM, Beijnen JH, Steeghs N, et al. Solubility and bioavailability improvement of pazopanib hydrochloride. Int J Pharm. 2018;544(1):181–90.

    Article  CAS  PubMed  Google Scholar 

  6. US Food and Drug administration, Votrient® Product Label. 2015 [Internet]. [cited 2024 Jan 11]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/022465s020lbl.pdf. Accessed 11 Jan 2024.

  7. Herbrink M, Nuijen B, Schellens JHM, Beijnen JH. Variability in bioavailability of small molecular tyrosine kinase inhibitors. Cancer Treat Rev [Internet]. 2015 [cited 2024 Jan 11];41:412–22. Available from: https://www.sciencedirect.com/science/article/pii/S030573721500050X. Accessed 11 Jan 2024.

  8. Choi SA, Park EJ, Lee JH, Min KA, Kim ST, Jang D-J, et al. Preparation and characterization of pazopanib hydrochloride-loaded four-component self-nanoemulsifying drug delivery systems preconcentrate for enhanced solubility and dissolution. Pharmaceutics [Internet]. 2022 [cited 2024 Jan 8];14:1875. Available from: https://www.mdpi.com/1999-4923/14/9/1875.

  9. Kumar D, Talegaonkar S, Bedi S, Dubey K. Design, development and evaluation of self-microemulsifying drug delivery system of pazopanib for enhanced dissolution rate and cytotoxic potential. J Drug Deliv Sci Technol [Internet]. 2023 [cited 2024 Jan 8];80:104181. Available from: https://www.sciencedirect.com/science/article/pii/S1773224723000333.

  10. Rai SK, Baidya D, Nangia AK. Salts, solvates and hydrates of the multi-kinase inhibitor drug pazopanib with hydroxybenzoic acids. CrystEngComm [Internet]. 2021 [cited 2024 Jan 8];23:5994–6011. Available from: https://pubs.rsc.org/en/content/articlelanding/2021/ce/d1ce00785h.

  11. Sirvi A, Debaje S, Guleria K, Sangamwar AT. Critical aspects involved in lipid dispersion and digestion: emphasis on in vitro models and factors influencing lipolysis of oral lipid based formulations. Adv Colloid Interface Sci. 2023;321:103028. Available from: https://www.sciencedirect.com/science/article/pii/S0001868623001951.

  12. Koehl NJ, Holm R, Kuentz M, Griffin BT. New insights into using lipid based suspensions for ‘Brick Dust’ molecules: case study of nilotinib. Pharm Res. 2019;36:56. https://doi.org/10.1007/s11095-019-2590-y. [cited 2022 Dec 26].

    Article  CAS  PubMed  Google Scholar 

  13. Nora G-I, Venkatasubramanian R, Strindberg S, Siqueira-Jørgensen SD, Pagano L, Romanski FS, et al. Combining lipid based drug delivery and amorphous solid dispersions for improved oral drug absorption of a poorly water-soluble drug. J Controlled Release [Internet]. 2022 [cited 2024 Jan 8];349:206–12. Available from: https://www.sciencedirect.com/science/article/pii/S0168365922003996.

  14. Williams HD, Ford L, Igonin A, Shan Z, Botti P, Morgen MM, et al. Unlocking the full potential of lipid-based formulations using lipophilic salt/ionic liquid forms. Adv Drug Deliv Rev [Internet]. 2019 [cited 2024 Jan 8];142:75–90. Available from: https://www.sciencedirect.com/science/article/pii/S0169409X19300584.

  15. Sahbaz Y, Nguyen TH, Ford L, McEvoy CL, Williams HD, Scammells PJ, et al. Ionic liquid forms of weakly acidic drugs in oral lipid formulations: preparation, characterization, in vitro digestion, and in vivo absorption studies. Mol Pharm. 2017;14:3669–83.

    Article  CAS  PubMed  Google Scholar 

  16. Khurana RK, Bansal AK, Beg S, Burrow AJ, Katare OP, Singh KK, et al. Enhancing biopharmaceutical attributes of phospholipid complex-loaded nanostructured lipidic carriers of mangiferin: systematic development, characterization and evaluation. Int J Pharm. 2017;518:289–306.

    Article  CAS  PubMed  Google Scholar 

  17. Huang J, Chen PX, Rogers MA, Wettig SD. Investigating the phospholipid effect on the bioaccessibility of rosmarinic acid-phospholipid complex through a dynamic gastrointestinal in vitro model. Pharmaceutics. 2019;11:156. Available from: https://www.mdpi.com/1999-4923/11/4/156.

  18. Mondal S, Sirvi A, Jadhav K, Sangamwar AT. Supersaturating lipid-based solid dispersion of atazanavir provides enhanced solubilization and supersaturation in the digestive aqueous phase. Int J Pharm [Internet]. 2023 [cited 2024 Jan 11];638:122919. Available from: https://www.sciencedirect.com/science/article/pii/S0378517323003393.

  19. Holm R, Kuentz M, Ilie-Spiridon A-R, Griffin BT. Lipid based formulations as supersaturating oral delivery systems: from current to future industrial applications. Eur J Pharm Sci [Internet]. 2023 [cited 2024 Jan 11];189:106556. Available from: https://www.sciencedirect.com/science/article/pii/S0928098723001860.

  20. Ford L, Tay E, Nguyen T-H, Williams HD, Benameur H, Scammells PJ, et al. API ionic liquids: probing the effect of counterion structure on physical form and lipid solubility. RSC Adv [Internet]. 2020 [cited 2023 Nov 1];10:12788–99. Available from: http://xlink.rsc.org/?DOI=D0RA00386G.

  21. Ditzinger F, Price DJ, Ilie A-R, Köhl NJ, Jankovic S, Tsakiridou G, et al. Lipophilicity and hydrophobicity considerations in bio-enabling oral formulations approaches – a PEARRL review. J Pharm Pharmacol. 2019;71:464–82. https://doi.org/10.1111/jphp.12984. [cited 2024 Jan 11].

    Article  CAS  PubMed  Google Scholar 

  22. Williams HD, Ford L, Han S, Tangso KJ, Lim S, Shackleford DM, et al. Enhancing the oral absorption of kinase inhibitors using lipophilic salts and lipid-based formulations. 2018;15(12):5678–96.

    CAS  Google Scholar 

  23. Sahbaz Y, Williams HD, Nguyen TH, Saunders J, Ford L, Charman SA, Scammells PJ, Porter CJ. Transformation of poorly water-soluble drugs into lipophilic ionic liquids enhances oral drug exposure from lipid based formulations. Mol Pharm. 2015;12(6):1980–91.

    Article  CAS  PubMed  Google Scholar 

  24. Sutar Y, Fulton SR, Paul S, Altamirano S, Mhatre S, Saeed H, et al. Docusate-based ionic liquids of anthelmintic benzimidazoles show improved pharmaceutical processability, lipid solubility, and in vitro activity against Cryptococcus neoformans. ACS Infect Dis. 2021;7(9):2637–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Saeed HK, Sutar Y, Patel P, Bhat R, Mallick S, Hatada AE, et al. Synthesis and characterization of lipophilic salts of metformin to improve its repurposing for cancer therapy. ACS Omega. 2021;6(4):2626–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kuentz M, Koehl NJ, Henze LJ, Keating JJ, De Vijlder T, Marx A, et al. Lipophilic salts and lipid-based formulations for bridging the food effect gap of venetoclax. J Pharm Sci. 2021;111(1):164–74.

    PubMed  Google Scholar 

  27. Kuche K, Bhargavi N, Dora CP, Jain S. Drug-phospholipid complex — a go through strategy for enhanced oral bioavailability. AAPS PharmSciTech. 2019;20(2):43.

    Article  CAS  PubMed  Google Scholar 

  28. Mandeep, Kaur S, Samal SK, Roy S, Sangamwar AT. Successful oral delivery of fexofenadine hydrochloride by improving permeability via phospholipid complexation. Eur J Pharm Sci. 2020;149:105338.

    Article  CAS  PubMed  Google Scholar 

  29. Zhang J, Peng Q, Shi S, Zhang Q, Sun X, Gong T, et al. Preparation, characterization, and in vivo evaluation of a self-nanoemulsifying drug delivery system (SNEDDS) loaded with morin-phospholipid complex. Int J Nanomedicine [Internet]. 2011 [cited 2024 Jan 11];6:3405–14. Available from: https://www.tandfonline.com/doi/abs/10.2147/IJN.S25824.

  30. Choi S-U, Kim MJ, Kim ST, Kim H-C, Cho KH, Park SY, et al. Development of self-microemulsifying drug delivery systems of poorly water-soluble pazopanib for improvement of oral absorption. Sci Adv Mater. 2020;12:152–60.

    Article  CAS  Google Scholar 

  31. Lai A, Sahbaz Y, Ford L, Nguyen TH, Haque S, Williams HD, et al. Stabilising disproportionation of lipophilic ionic liquid salts in lipid-based formulations. Int J Pharm. 2021;597:1–37.

    Article  Google Scholar 

  32. Dora CP, Kushwah V, Katiyar SS, Kumar P, Pillay V, Suresh S, et al. Improved oral bioavailability and therapeutic efficacy of erlotinib through molecular complexation with phospholipid. Int J Pharm. 2017;534:1–13.

    Article  CAS  PubMed  Google Scholar 

  33. Sirvi A, Kuche K, Chaudhari D, Ghadi R, Date T, Katiyar SS, et al. Supersaturable self-emulsifying drug delivery system: a strategy for improving the loading and oral bioavailability of quercetin. J Drug Deliv Sci Technol [Internet]. 2022 [cited 2022 Oct 27];71:103289. Available from: https://www.sciencedirect.com/science/article/pii/S177322472200199X.

  34. Mosgaard MD, Sassene P, Mu H, Rades T, Müllertz A. European Journal of Pharmaceutics and biopharmaceutics development of a high-throughput in vitro intestinal lipolysis model for rapid screening of lipid-based drug delivery systems. Eur J Pharm Biopharm. 2015;94:493–500.

    Article  CAS  PubMed  Google Scholar 

  35. Kilic M, Dressman J. A simplified method to screen for in-vivo performance of oral lipid formulations. J Pharm Pharmacol. 2014;66:615–23. https://doi.org/10.1111/jphp.12182. [cited 2024 Jan 8].

    Article  CAS  PubMed  Google Scholar 

  36. Williams HD, Ford L, Lim S, Han S, Baumann J, Sullivan H, et al. Transformation of biopharmaceutical classi fi cation system class I and III drugs into ionic liquids and lipophilic salts for enhanced developability using lipid formulations. J Pharm Sci. 2018;107:203–16.

    Article  CAS  PubMed  Google Scholar 

  37. Telange DR, Sohail NK, Hemke AT, Kharkar PS, Pethe AM. Phospholipid complex-loaded self-assembled phytosomal soft nanoparticles: evidence of enhanced solubility, dissolution rate, ex vivo permeability, oral bioavailability, and antioxidant potential of mangiferin. Drug Deliv Transl Res. 2021;11:1056–83.

    Article  CAS  PubMed  Google Scholar 

  38. Chakravarti RK, Kaur S, Samal SK, Kashyap MC, Sangamwar AT. Combination of phospholipid complex and matrix dispersion. AAPS PharmSciTech. 2021;22:189. https://doi.org/10.1208/s12249-021-02067-x. [cited 2024 Jan 8].

    Article  CAS  PubMed  Google Scholar 

  39. Tay E, Nguyen T-H, Ford L, Williams HD, Benameur H, Scammells PJ, et al. Ionic liquid forms of the antimalarial lumefantrine in combination with LFCS type IIIB lipid-based formulations preferentially increase lipid solubility, in vitro solubilization behavior and in vivo exposure. Pharmaceutics [Internet]. 2020 [cited 2024 Jan 9];12:17. Available from: https://www.mdpi.com/1999-4923/12/1/17.

  40. Bannow J, Yorulmaz Y, Löbmann K, Müllertz A, Rades T. Improving the drug load and in vitro performance of supersaturated self-nanoemulsifying drug delivery systems (super-SNEDDS) using polymeric precipitation inhibitors. Int J Pharm. 2020;575:118960.

    Article  CAS  PubMed  Google Scholar 

  41. Morgen M, Saxena A, Chen X-Q, Miller W, Nkansah R, Goodwin A, et al. Lipophilic salts of poorly soluble compounds to enable high-dose lipidic SEDDS formulations in drug discovery. Eur J Pharm Biopharm [Internet]. 2017 [cited 2022 Nov 22];117:212–23. Available from: https://www.sciencedirect.com/science/article/pii/S0939641116310402.

  42. Kumar D, Talegaonkar S, Bedi S, Dubey K. Journal of Drug Delivery Science and Technology Design, development and evaluation of self-microemulsifying drug delivery system of pazopanib for enhanced dissolution rate and cytotoxic potential. J Drug Deliv Sci Technol. 2023;80:104181.

    Article  CAS  Google Scholar 

  43. Tanaka Y, Tay E, Nguyen T-H, Porter CJH. Quantifying in vivo luminal drug solubilization -supersaturation-precipitation profiles to explain the performance of lipid based formulations. Pharm Res. 2020;37:47. https://doi.org/10.1007/s11095-020-2762-9. [cited 2022 Nov 18].

    Article  CAS  PubMed  Google Scholar 

  44. Stillhart C, Imanidis G, Kuentz M. Insights into drug precipitation kinetics during in vitro digestion of a lipid-based drug delivery system using in-line Raman spectroscopy and mathematical modeling. Pharm Res. 2013;30:3114–30. https://doi.org/10.1007/s11095-013-0999-2. [cited 2023 Jun 21].

    Article  CAS  PubMed  Google Scholar 

  45. Anby MU, Williams HD, McIntosh M, Benameur H, Edwards GA, Pouton CW, et al. Lipid digestion as a trigger for supersaturation: evaluation of the impact of supersaturation stabilization on the in vitro and in vivo performance of self-emulsifying drug delivery systems. Mol Pharm. 2012;9:2063–79. https://doi.org/10.1021/mp300164u. [cited 2024 Jan 8].

    Article  CAS  PubMed  Google Scholar 

  46. Ilie A-R, Griffin BT, Brandl M, Bauer-Brandl A, Jacobsen A-C, Vertzoni M, et al. Exploring impact of supersaturated lipid-based drug delivery systems of celecoxib on in vitro permeation across PermeapadⓇ membrane and in vivo absorption. Eur J Pharm Sci [Internet]. 2020 [cited 2022 Oct 29];152:105452. Available from: https://www.sciencedirect.com/science/article/pii/S0928098720302414.

  47. Gamboa A, Schüßler N, Soto-Bustamante E, Romero-Hasler P, Meinel L, Morales JO. Delivery of ionizable hydrophilic drugs based on pharmaceutical formulation of ion pairs and ionic liquids. Eur J Pharm Biopharm [Internet]. 2020 [cited 2024 Jan 9];156:203–18. Available from: https://www.sciencedirect.com/science/article/pii/S093964112030285X.

  48. Li X, Ma N, Zhang L, Ling G, Zhang P. Applications of choline-based ionic liquids in drug delivery. Int J Pharm [Internet]. 2022 [cited 2024 Jan 11];612:121366. Available from: https://www.sciencedirect.com/science/article/pii/S0378517321011728.

  49. Holm R. Bridging the gaps between academic research and industrial product developments of lipid-based formulations. Adv Drug Deliv Rev [Internet]. 2019 [cited 2024 Jan 11];142:118–27. Available from: https://www.sciencedirect.com/science/article/pii/S0169409X19300122.

  50. Chen F-J, Etzler FM, Ubben J, Birch A, Zhong L, Schwabe R, et al. Effects of lipophilic components on the compatibility of lipid-based formulations with hard gelatin capsules. J Pharm Sci [Internet]. 2010 [cited 2024 Jan 11];99:128–41. Available from: https://www.sciencedirect.com/science/article/pii/S0022354916303744.

  51. Bannow J, Yorulmaz Y, Löbmann K, Müllertz A, Rades T. Improving the drug load and in vitro performance of supersaturated self-nanoemulsifying drug delivery systems (super-SNEDDS) using polymeric precipitation inhibitors. Int J Pharm [Internet]. 2020 [cited 2024 Jan 8];575:118960. Available from: https://www.sciencedirect.com/science/article/pii/S0378517319310051.

  52. Koehl NJ, Henze LJ, Bennett-Lenane H, Faisal W, Price DJ, Holm R, et al. In silico, in vitro, and in vivo evaluation of precipitation inhibitors in supersaturated lipid-based formulations of venetoclax. Mol Pharm. 2021;18:2174–88. https://doi.org/10.1021/acs.molpharmaceut.0c00645. [cited 2024 Jan 11].

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are thankful to the Director of NIPER S.A.S. Nagar for providing the infrastructure and amenities required to undertake the experimental investigations.

Author information

Authors and Affiliations

Authors

Contributions

KJ: conceptualization, methodology, investigation, visualization, writing—original draft. AS: conceptualization, investigation, writing—original draft, writing—review and editing. AJ: methodology, data curation. MCK: methodology (pharmacokinetic study) ATS: conceptualization, funding acquisition, project administration, resources, writing—review, and editing.

Corresponding author

Correspondence to Abhay T. Sangamwar.

Ethics declarations

Ethical Approval

All aspects of animal handling, care, and experimentation adhere to the ARRIVE guidelines and comply with the UK Animals (Scientific Procedures) Act, 1986, along with associated guidelines and the EU Directive 2010/63/EU for animal experiments. The protocol necessary for conducting a pharmacokinetic study on laboratory animals received approval from the Institutional Animal Ethics Committee, NIPER S.A.S. Nagar, India.

Conflict of Interest

The authors declare no competing interests.

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 (DOCX 8886 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jadhav, K., Sirvi, A., Janjal, A. et al. Utilization of Lipophilic Salt and Phospholipid Complex in Lipid-Based Formulations to Modulate Drug Loading and Oral Bioavailability of Pazopanib. AAPS PharmSciTech 25, 59 (2024). https://doi.org/10.1208/s12249-024-02780-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12249-024-02780-3

Keywords

Navigation