Skip to main content
Log in

MALDI imaging facilitates new topical drug development process by determining quantitative skin distribution profiles

  • Research Paper
  • Published:
Analytical and Bioanalytical Chemistry Aims and scope Submit manuscript

Abstract

Generation of skin distribution profiles and reliable determination of drug molecule concentration in the target region are crucial during the development process of topical products for treatment of skin diseases like psoriasis and atopic dermatitis. Imaging techniques like mass spectrometric imaging (MSI) offer sufficient spatial resolution to generate meaningful distribution profiles of a drug molecule across a skin section. In this study, we use matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to generate quantitative skin distribution profiles based on tissue extinction coefficient (TEC) determinations of four different molecules in cross sections of human skin explants after topical administration. The four drug molecules: roflumilast, tofacitinib, ruxolitinib, and LEO 29102 have different physicochemical properties. In addition, tofacitinib was administrated in two different formulations. The study reveals that with MALDI-MSI, we were able to observe differences in penetration profiles for both the four drug molecules and the two formulations and thereby demonstrate its applicability as a screening tool when developing a topical drug product. Furthermore, the study reveals that the sensitivity of the MALDI-MSI techniques appears to be inversely correlated to the drug molecules’ ability to bind to the surrounding tissues, which can be estimated by their Log D values.

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.

Institutional subscriptions

Chart 1
Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Wilsmann-Theis D, Hagemann T, Jordan J, Bieber T, Novak N. Facing psoriasis and atopic dermatitis: are there more similarities or more differences? Eur J Dermatology. 2008;18:172–80. https://doi.org/10.1684/ejd.2008.0357.

    Google Scholar 

  2. Schon MP, Boehncke WH. Psoriasis. N Engl J Med. 2005;352:1899–912.

    Article  CAS  Google Scholar 

  3. Menter A, Gottlieb A, Feldman SR, Van Voorhees AS, Leonardi CL, Gordon KB, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. Section 1. Overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol. 2008;58:826–50. https://doi.org/10.1016/j.jaad.2008.02.039.

  4. Hanifin J, Rajka G. Diagnostic features of atopic dermatitis. Acta Dermatovener. 1980;92:44–7.

    Google Scholar 

  5. Lewis-Jones S. Quality of life and childhood atopic dermatitis: the misery of living with childhood eczema. Int J Clin Pract. 2006;60:984–92. https://doi.org/10.1111/j.1742-1241.2006.01047.x.

    Article  CAS  Google Scholar 

  6. Ständer S, Steinhoff M. Pathophysiology of pruritus in atopic dermatitis: an overview. Exp Dermatol. 2002;11:12–24. https://doi.org/10.1034/j.1600-0625.2002.110102.x.

    Article  Google Scholar 

  7. Menter A, Korman NJ, Elmets CA, Feldman SR, Gelfand JM, Gordon KB, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. Section 3. Guidelines of care for the management and treatment of psoriasis with topical therapies. J Am Acad Dermatol. 2009;60:643–59. https://doi.org/10.1016/j.jaad.2008.12.032.

  8. Thaçi D, Salgo R. Malignancy concerns of topical calcineurin inhibitors for atopic dermatitis: facts and controversies. Clin Dermatol. 2010;28:52–6. https://doi.org/10.1016/j.clindermatol.2009.04.001.

    Article  Google Scholar 

  9. Leite-Silva V, de Almeida M, Fradin A, Grice JE, Roberts MS. Delivery of drugs applied topically to the skin. Expert Rev Dermatol. 2012;7:383–97. https://doi.org/10.1586/edm.12.32.

    Article  CAS  Google Scholar 

  10. Feingold KR. Thematic review series: skin lipids. The role of epidermal lipids in cutaneous permeability barrier homeostasis. J Lipid Res. 2007;48:2531–46. https://doi.org/10.1194/jlr.R700013-JLR200.

    Article  CAS  Google Scholar 

  11. Michaels AS, Chandrasekaran SK, Shaw JE. Drug permeation through human skin: theory and invitro experimental measurement. AICHE J. 1975;21:985–96. https://doi.org/10.1002/aic.690210522.

    Article  CAS  Google Scholar 

  12. Elias PM. Epidermal lipids, barrier function, and desquamation. J Invest Dermatol. 1983;80:44s–9s. https://doi.org/10.1111/1523-1747.ep12537108.

    Article  CAS  Google Scholar 

  13. Van Smeden J, Janssens M, Gooris GS, Bouwstra JA. The important role of stratum corneum lipids for the cutaneous barrier function. Biochim Biophys Acta - Mol Cell Biol Lipids. 2014;1841:295–313. https://doi.org/10.1016/j.bbalip.2013.11.00.

    Article  Google Scholar 

  14. Hadgraft J, Lane ME. Skin permeation: the years of enlightenment. Int J Pharm. 2005;305:2–12. https://doi.org/10.1016/j.ijpharm.2005.07.014.

    Article  CAS  Google Scholar 

  15. Marks R. The stratum corneum barrier: the final frontier. J Nutr. 2004;134:2017S–21S.

    Article  CAS  Google Scholar 

  16. Vickers CH. Existence of reservoir in the stratum corneum: experimental proof. Arch Dermatol. 1963;88:20–3.

    Article  CAS  Google Scholar 

  17. Trommer H, Neubert RHH. Overcoming the stratum Corneum: the modulation of skin penetration a review structure of the stratum corneum and drug options to overcome the barrier. Skin Pharmacol Physiol. 2006;19:106–21.

    Article  CAS  Google Scholar 

  18. Harrison JE, Watkinson AC, Green DM, Hadgraft J, Brain K. The relative effect of Azone(R) and Transcutol(R) on permeant diffusivity and solubility in human stratum corneum. Pharm Res. 1996;13:542–6. https://doi.org/10.1023/A:1016037803128.

    Article  CAS  Google Scholar 

  19. Williams A, Barry B. Penetration enhancers. Adv Drug Deliv Rev. 2012;64:128–37. https://doi.org/10.1016/j.addr.2012.09.032.

    Article  Google Scholar 

  20. Franz TJ. Percutaneous absorption. On the relevance of in vitro data. J Invest Dermatol. 1975;64:190–5. https://doi.org/10.1111/1523-1747.ep12533356.

    Article  CAS  Google Scholar 

  21. Kligman AM, Chirstophers E. preparation of isolated sheets of human stratum corneum. Arch Dermatol. 1963;88:702–5.

    Article  CAS  Google Scholar 

  22. Lind M, Nielsen KT, Schefe LH, Nørremark K, Eriksson AH, Norsgaard H, et al. Supersaturation of calcipotriene and betamethasone dipropionate in a novel aerosol foam formulation for topical treatment of psoriasis provides enhanced bioavailability of the active ingredients. Dermatol Ther (Heidelb). 2016;6:413–25. https://doi.org/10.1007/s13555-016-0125-6.

  23. Alvarez-Roman R, Naik A, Kalia YN, Fessi H, Guy RH. Visualization of skin penetration using confocal laser scanning microscopy. Eur J Pharm Biopharm. 2004;58:301–16.

    Article  CAS  Google Scholar 

  24. Mendelsohn R, Flach CR, Moore DJ. Determination of molecular conformation and permeation in skin via IR spectroscopy, microscopy, and imaging. Biochim Biophys Acta Biomembr. 2006;1758:923–33. https://doi.org/10.1016/j.bbamem.2006.04.009.

    Article  CAS  Google Scholar 

  25. Simona Mura, Maria Manconi, Anna Maria Fadda, Maria Chiara Sala, Jacopo Perricc, Elena Pini CS (2012) Penetration enhancer-containing vesicles (PEVs) as carriers for cutaneous delivery of minoxidil: in vitro evaluation of drug permeation by infrared spectroscopy Read More: http://informahealthcare.com/doi/abs/10.3109/10837450.2012.685661. Pharm Dev Technol 18:1–7. https://doi.org/10.3109/10837450.2012.685661

  26. Mao G, Flach CR, Mendelsohn R, Walters RM. Imaging the distribution of sodium dodecyl sulfate in skin by confocal Raman and infrared microspectroscopy. Pharm Res. 2012;29:2189–201.

    Article  CAS  Google Scholar 

  27. Freudiger CW, Min W, Saar BG, Lu S, Holtom GR, He C, et al. Label-free biomedical imaging with high sensitivity by stimulated raman scattering microscopy. Science. 2008;322(80):1857–61.

  28. Caprioli RM, Farmer TB, Gile J. Molecular imaging of biological samples: localization of peptides and proteins using MALDI-TOF MS. Anal Chem. 1997;69:4751–60. https://doi.org/10.1021/Ac970888i.

    Article  CAS  Google Scholar 

  29. Bunch J, Clench MR, Richards DS. Determination of pharmaceutical compounds in skin by imageing matrix-assisted laser desorption/ionisation mass spectrometry. Rapid Commun Mass Spectrom. 2004;18:3051–60.

    Article  CAS  Google Scholar 

  30. Hart PJ, Francese S, Claude E, Woodroofe MN, Clench MR. MALDI-MS imaging of lipids in ex vivo human skin. Anal Bioanal Chem. 2011;401:115–25. https://doi.org/10.1007/s00216-011-5090-4.

    Article  CAS  Google Scholar 

  31. Enthaler B, Pruns JK, Wessel S, Rapp C, Fischer M, Wittern KP. Improved sample preparation for MALDI-MSI of endogenous compounds in skin tissue sections and mapping of exogenous active compounds subsequent to ex-vivo skin penetration. Anal Bioanal Chem. 2012;402:1159–67. https://doi.org/10.1007/s00216-011-5562-6.

    Article  CAS  Google Scholar 

  32. Hunt DW, Winters GC, Brownsey RW, Kulpa JE, Gilliland KL, Thiboutot DM, Hofland HE (2017) Inhibition of sebum production with the acetyl coenzyme a carboxylase inhibitor olumacostat glasaretil. J Invest Dermatol. https://doi.org/10.1016/j.jid.2016.12.031.

  33. Sorensen IS, Janfelt C, Nielsen MMB, Mortensen RW, Knudsen NO, Eriksson AH, et al. Combination of MALDI-MSI and cassette dosing for evaluation of drug distribution in human skin explant. Anal Bioanal Chem. 2017;409:4993–5005. https://doi.org/10.1007/s00216-017-0443-2.

  34. Judd AM, Scurr DJ, Heylings JR, Wan KW, Moss GP. Distribution and visualisation of chlorhexidine within the skin using ToF-SIMS: a potential platform for the design of more efficacious skin antiseptic formulations. Pharm Res. 2013;30:1896–905. https://doi.org/10.1007/s11095-013-1032-5.

    Article  CAS  Google Scholar 

  35. Čižinauskas V, Elie N, Brunelle A, Briedis V. Fatty acids penetration into human skin ex vivo : a TOF-SIMS analysis approach. Biointerphases. 2017;12:11003. https://doi.org/10.1116/1.4977941.

    Article  Google Scholar 

  36. Sjövall P, Greve TM, Clausen SK, Moller K, Eirefelt S, Johansson B, et al. Imaging of distribution of topically applied drug molecules in mouse skin by combination of time-of-flight secondary ion mass spectrometry and scanning electron microscopy. Anal Chem. 2014;86:3443–52. https://doi.org/10.1021/ac403924w.

  37. Stauber J. Quantitation by MS imaging: needs and challenges in pharmaceuticals. Bioanalysis. 2012;4:2095–8.

    Article  CAS  Google Scholar 

  38. Pirman DA, Reich RF, Kiss A, Heeren RMA, Yost RA. Quantitative MALDI tandem mass spectrometric imaging of cocaine from brain tissue with a deuterated internal standard. Anal Chem. 2013;85:1081–9. https://doi.org/10.1021/ac302960j.

    Article  CAS  Google Scholar 

  39. Turker SD, Dunn WB, Wilkie J. MALDI MS of drugs: profiling, imaging and steps towards quantitative analysis. Appl Spectrosc Rev. 2016;4928:00–0. https://doi.org/10.1080/05704928.2016.1207659.

  40. Takai N, Tanaka Y, Watanabe A, Saji H. Quantitative imaging of a therapeutic peptide in biological tissue sections by MALDI MS. Bioanalysis. 2013;5:603–12. https://doi.org/10.4155/bio.13.13.

    Article  CAS  Google Scholar 

  41. Hamm G, Bonnel D, Legouffe R, Pamelard F, Delbos JM, Bouzom F, et al. Quantitative mass spectrometry imaging of propranolol and olanzapine using tissue extinction calculation as normalization factor. J Proteome. 2012;75:4952–61. https://doi.org/10.1016/j.jprot.2012.07.035.

  42. Stoeckli M, Staab D, Schweitzer A. Compound and metabolite distribution measured by MALDI mass spectrometric imaging in whole-body tissue sections. Int J Mass Spectrom. 2007;260:195–202. https://doi.org/10.1016/j.ijms.2006.10.007.

    Article  CAS  Google Scholar 

  43. Felding J, Sørensen MD, Poulsen TD, Larsen J, Andersson C, Refer P, et al. Discovery and early clinical development of 2-{6-[2-(3,5-Dichloro-4-pyridyl)acetyl]-2,3-dimethoxyphenoxy}-N-propylacetamide (LEO 29102), a soft-drug inhibitor of phosphodiesterase 4 for topical treatment of atopic dermatitis. J Med Chem. 2014;57:5893–903.

  44. Boswell-Smith V, Spina D. PDE4 inhibitors as potential therapeutic agents in the treatment of COPD-focus on roflumilast. Int J Chron Obs Pulmon Dis. 2007;2:121–9.

    CAS  Google Scholar 

  45. Garnock-Jones KP. Roflumilast: a review in COPD. Drugs. 2015;75:1645–56. https://doi.org/10.1007/s40265-015-0463-1.

    Article  CAS  Google Scholar 

  46. Tofacitinib. Drugs R D. 2010;10:271–84.

  47. Bachelez H, Van De Kerkhof PCM, Strohal R, Kubanov A, Valenzuela F, Lee JH, et al. Tofacitinib versus etanercept or placebo in moderate-to-severe chronic plaque psoriasis: a phase 3 randomised non-inferiority trial. Lancet. 2015;386:552–61. https://doi.org/10.1016/S0140-6736(14)62113-9.

  48. Mesa RA, Yasothan U, Kirkpatrick P. Ruxolitinib. Nat Rev Drug Discov. 2012;11(2):103–4. https://doi.org/10.1038/nrd3652.

    Article  CAS  Google Scholar 

  49. Kofoed K, Skov L, Zachariae C. New drugs and treatment targets in psoriasis. Acta Derm Venereol. 2015;95:133–9. https://doi.org/10.2340/00015555-1931.

    Article  Google Scholar 

  50. Magnusson BM, Anissimov YG, Cross SE, Roberts MS. Molecular size as the main determinant of solute maximum flux across the skin. J Invest Dermatol. 2004;122:993–9. https://doi.org/10.1111/j.0022-202X.2004.22413.x.

    Article  CAS  Google Scholar 

  51. Potts RO, Guy RH. A predictive algorithm for skin permeability: the effects of molecular size and hydrogen bond activity. Pharm Res An Off J Am Assoc Pharm Sci. 1995;12:1628–33. https://doi.org/10.1023/A:1016236932339.

    CAS  Google Scholar 

  52. Magnusson BM, Cross SE, Winckle G, Roberts MS. Percutaneous absorption of steroids: determination of in vitro permeability and tissue reservoir characteristics in human skin layers. Skin Pharmacol Physiol. 2006;19:336–42. https://doi.org/10.1159/000095254.

    Article  CAS  Google Scholar 

  53. Zhang Q, Li P, Roberts MS. Maximum transepidermal flux for similar size phenolic compounds is enhanced by solvent uptake into the skin. J Control Release. 2011;154:50–7. https://doi.org/10.1016/j.jconrel.2011.04.018.

    Article  CAS  Google Scholar 

  54. Zhang Q, Li P, Liu D, Roberts MS. Effect of vehicles on the maximum transepidermal flux of similar size phenolic compounds. Pharm Res. 2012;30:1–9. https://doi.org/10.1007/s11095-012-0846-x.

    Google Scholar 

  55. Sanagi MM, Ling SL, Nasir Z, Hermawan D, Wan Ibrahim WA, Naim AA. Comparison of signal-to-noise, blank determination, and linear regression methods for the estimation of detection and quantification limits for volatile organic compounds by gas chromatography. J AOAC Int. 2009;92:1833–8.

    CAS  Google Scholar 

  56. Laznicek M, Laznickova A. The effect of lipophilicity on the protein binding and blood cell uptake of some acidic drugs. J Pharm Biomed Anal. 1995;13:823–8.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge Tina Leonhardt Hjort for the execution of the drug penetration study, Liselotte Saustrup Kirk for the UHPLC-MS/MS analysis of the tissue samples, and Lotte Koue for the help with the production of the formulations. We also acknowledge Torsten Kåre Askland, Edma Edge Fontaine, and Mathieu Gaudin for helping with the proofreading.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kim T. Nielsen.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Informed consent

The study involved anonymized human skin samples from abdominoplasty surgery acquired from Biopredic International, France, upon written informed consent from the donor.

Ethical approval

According to the Danish Committee Act section 14.3, anonymous human biological material used in health research projects shall not seek ethical approval as long as the collected human biological material has been collected lawfully in the country of origin which is the case. The supplier of the human biological material holds a permit granted by the French Ministry of Higher Education and Research for the acquisition, transformation, sales, and export of human biological material to be used in research, and is furthermore in compliance with the French law CSP1245-2.

Electronic supplementary material

ESM 1

(PDF 220 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bonnel, D., Legouffe, R., Eriksson, A.H. et al. MALDI imaging facilitates new topical drug development process by determining quantitative skin distribution profiles. Anal Bioanal Chem 410, 2815–2828 (2018). https://doi.org/10.1007/s00216-018-0964-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00216-018-0964-3

Keywords

Navigation