Skip to main content

Chapter 18: Design of Clinical In-Use Studies

  • Chapter
  • First Online:
Book cover Development of Biopharmaceutical Drug-Device Products

Abstract

For most parenteral biologics, in-use manipulation of drug product during the aseptic dose solution preparation and administration process is inevitable. The in-use product handling procedures, and compatibility issues with administration components used in the clinical setting, can potentially impact protein stability. Therefore, it becomes imperative to define dose solution preparation, handling, and administration procedures that ensure the quality of the product solution administered to patients.

The general clinical in-use study design recommendations and case studies presented in this chapter highlight the commonly observed stability issues for parenteral intravenous protein products. This information can be used by pharmaceutical scientists as a practical guide to design and execute robust in-use studies through the development cycle of a drug product candidate.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 189.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 249.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Gervasi V, Dall AR, Cullen S, Mccoy T, Vucen S, Crean A. Parenteral protein formulations: an overview of approved products within the European Union. Eur J Pharm Biopharm. 2018;131:8–24.

    Article  CAS  PubMed  Google Scholar 

  2. Mitragotri S, Burke PA, Langer R. Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat Rev Drug Discov. 2014;13(9):655–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Geigert J. The challenge of CMC regulatory compliance for biopharmaceuticals and other biologics. 2nd ed: Springer; 2013.

    Google Scholar 

  4. ICH Harmonised Tripartite Guideline Q8 (R2) (2009): Pharmaceutical Development 1-18. https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q8_R1/Step4/Q8_R2_Guideline.pdf. Accessed November 26, 2018.

  5. Guidance for Industry: dosage and administration section of labeling for human prescription drug and biological products — content and format. https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm075066.pdf. Accessed March 1, 2019.

  6. FDA 21 CFR 201.57(c)(3): dosage and administration. https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/cfrsearch.cfm?fr=201.57. Accessed March 1, 2019.

  7. Regulation (EU) No. 536/2014 of the European Parliament and of the Council of 16 April 2014 on clinical trials on medicinal products for human use, and repealing Directive 2001/20/EC. https://ec.europa.eu/health/sites/health/files/files/eudralex/vol-1/reg_2014_536/reg_2014_536_en.pdf. Accessed March 7, 2019.

  8. ICH Harmonised Tripartite Guideline Q1A(R2): Stability testing of new drug substances and products. https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q1A_R2/Step4/Q1A_R2__Guideline.pdf Accessed February 12, 2019.

  9. ICH Harmonised Tripartite Guideline Q5C: Stability testing of biotechnological/biological products. https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q5C/Step4/Q5C_Guideline.pdf. Accessed March 7, 2019.

  10. EMA/CHMP/QWP/545525/2017: guideline on the requirements for the chemical and pharmaceutical quality documentation concerning investigational medicinal products in clinical trials. https://www.ema.europa.eu/documents/scientific-guideline/guideline-requirements-chemical-pharmaceutical-quality-documentation-concerning-investigational_en-0.pdf. Accessed November 28, 2018.

  11. CPMP/QWP/2934/99: note for guidance on In-use stability testing of human medicinal products. https://www.ema.europa.eu/en/documents/scientific-guideline/note-guidance-use-stability-testing-human-medicinal-products_en.pdf. Accessed March 7, 2019.

  12. CPMP/QWP/159/96: note for guidance on maximum shelf-life for sterile products after first opening or following reconstitution. https://www.ema.europa.eu/en/documents/scientific-guideline/note-guidance-maximum-shelf-life-sterile-products-human-use-after-first-opening-following_en.pdf. Accessed March 7, 2019.

  13. WHO Technical Report Series, No. 953, 2009. Stability testing of active pharmaceutical ingredients and finished pharmaceutical products. http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q1F/Stability_Guideline_WHO.pdf. Accessed March 7, 2019.

  14. USP<797> Pharmaceutical Compounding-Sterile Preparations.

    Google Scholar 

  15. USP<51> Antimicrobial Effectiveness Testing.

    Google Scholar 

  16. Ricci MS, Frazier M, Moore J, Cromwell M, Galush WJ, Patel AR, Adler M, Altenburger U, Grauschopf U, Goldbach P, Fast JL, Kramer I, Mahler HC. In-use physicochemical and microbiological stability of biological parenteral products. Am J Health-Syst Pharm. 2015;72(5):396–407.

    Article  PubMed  Google Scholar 

  17. Nejadnik MR, Randlph TW, Volkin DB, Schoneich C, Carpenter J, Crommelin Daan JA, Jiskoot W. Postproduction handling and administration of protein pharmaceuticals and potential instability issues. J Pharm Sci. 2018;107:2013–9.

    Article  CAS  PubMed  Google Scholar 

  18. Vlieland ND, Gardarsdottir H, Bouvy ML, Egberts TC, van den Bemt BJ. The majority of patients do not store their biologic disease-modifying antirheumatic drugs within the recommended temperature range. Rheumatology (Oxford). 2016;55(4):704–9.

    Article  Google Scholar 

  19. Jiskoot W, Nejadnik MR, Sediq AS. Potential issues with the handling of biologicals in a hospital. J Pharm Sci. 2017;106(6):1688–9.

    Article  CAS  PubMed  Google Scholar 

  20. Sharma M, Cheung JK, Debbara A, Pertersen J. Intravenous admixture compatibility for sterile products: challenges and regulatory guidance. In: Kolhe P, Shah M, Rathore N, editors. Sterile product development – formulation, process and regulatory considerations, AAPS advances in the pharmaceutical sciences series 6. New York: Springer; 2013. p. 461–74.

    Chapter  Google Scholar 

  21. US Department of Health and Human Services and Drug Administration Guidance for Industry (2010): Dosage and adminitration section of labeling for human prescription drug and biological products—content and format. https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm075066.pdf. Accessed March 1, 2019.

  22. Sedykh SE, Prinz VV, Buneva VN, Nevinsky GA. Bispecific antibodies: design, therapy, perspectives. Drug Des Devel Ther. 2018;12:195–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Reddi AS. Intravenous fluids: composition and indications. In book: Fluid, electrolyte and acid-base disorders. Springer. 2018. https://rd.springer.com/book/10.1007/978-1-4614-9083-8. Accessed March 7, 2019.

  24. Wang W. Instability, stabilization, and formulation of liquid protein pharmaceuticals. Int J Pharm. 1999;185(2):129–88.

    Article  CAS  PubMed  Google Scholar 

  25. Khan TA, Mahler HC, Kishore RS. Key interactions of surfactants in therapeutic protein formulations: a review. Eur J Pharm Biopharm. 2015;97(A):60–7.

    Article  CAS  PubMed  Google Scholar 

  26. Minning M, Liu J, Li TS, Holcomb RE. Rational design of liquid formulations of proteins. Adv Protein Chem Struct Biol. 2018;112:1–59.

    Article  CAS  Google Scholar 

  27. Kamerzell TJ, Esfandiary R, Joshi SB, Middaugh CR, Volkin DB. Protein-excipient interactions: mechanisms and biophysical characterization applied to protein formulation development. Adv Drug Deliv Rev. 2011;63(13):1118–59.

    Article  CAS  PubMed  Google Scholar 

  28. Hawe A, Friess W. Formulation development for hydrophobic therapeutic proteins. Pharm Dev Technol. 2007;12(3):223–37.

    Article  CAS  PubMed  Google Scholar 

  29. Avastin (bevacizumab)-FDA. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125085s319lbl.pdf. Accessed March 8, 2019.

  30. Herceptin-Product Information-Therapeutic Goods Administration. https://www.tga.gov.au/sites/default/files/auspar-trastuzumab-121217-pi.pdf. Accessed March 8, 2019. Springer, New York, NY.

  31. Fischer S, Hoemschemeyer J, Mahler HC. Glycation during storage and administration of monoclonal antibody formulations. Eur J Pharm Biopharm. 2008;70(1):42–50.

    Article  CAS  PubMed  Google Scholar 

  32. Zheng SY, Adams M, Mantri RV. An approach to mitigate particle formation on the dilution of a monoclonal antibody drug product in an IV administration fluid. J Pharm Sci. 2016;105(3):1349–50.

    Article  CAS  PubMed  Google Scholar 

  33. Giannos SA, Kraft ER, Zhao ZY, Merkley KH, Cai JY. Formulation stabilization and disaggregation of bevacizumab, ranibizumab and aflibercept in dilute solutions. Pharm Res. 2018;35(4):78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Preparing BLINCYTO®. https://www.blincyto.com/hcp/rr/dosing/preparation Accessed September 22, 2018.

  35. ISMP Canada. Managing overfill during preparation and delivery of intravenous medications. ISMP Canada Safety Bull. 2013;13(7):1–5. https://www.ismp-canada.org/download/safetyBulletins/2013/ISMPCSB2013-07_ManagingOverfillIntravenousMedications.pdf. Accessed March 7, 2019.

    Google Scholar 

  36. Syringe pull-back: a stubborn but unsafe drug prep method. 2015. https://www.pharmacypracticenews.com/Clinical/Article/09-15/Syringe-Pull-Back-A-Stubborn-But-Unsafe-Drug-Prep-Method/33437. Accessed March 7, 2019.

  37. Post-production IV admixture checks less than ideal. http://www.baxtermedicationdeliveryproducts.com/pdf/111835_ISMPPost-ProductionIVAdmixtureChecksLessThanIdeal_ArticleReprint_FINAL.pdf. Accessed March 7, 2019.

  38. Rios C, Vialet R, Dosmas C, Loundou A, Michel F. Comparing three methods for dilution accuracy of intravenous preparations. J Perianesth Nurs. 2016;31(2):113–7.

    Article  PubMed  Google Scholar 

  39. Stucki C, Sautter AM, Wolff A, Fleury-Souverain S, Bonnabry P. Accuracy of preparation of i.v. medication syringes for anesthesiology. Am J Health Syst Pharm. 2013;70(2):137–42.

    Article  CAS  PubMed  Google Scholar 

  40. Wheeler DW, Degnan BA, Sehmi JS, Burnstein RM, Menon DK, Gupta AK. Variability in the concentrations of intravenous drug infusions prepared in a critical care unit. Intensive Care Med. 2008;34(8):1441–7.

    Article  PubMed  Google Scholar 

  41. Massoomi F, PharmD, FASHP. The evolution of the CSTD. Oncol Safety. 2015;12(2):1.

    Google Scholar 

  42. Bee JS, Randolph TW, Carpenter JF, Bishop SM, Dimitrova MN. Effect of surfaces and leachable on the stability of biopharmaceuticals. J Pharm Sci. 2011;100(10):4158–70.

    Article  CAS  PubMed  Google Scholar 

  43. Fujita R, Nagatoishi S, Adachi S, Nishioka H, Ninomiya H, Kaya T, Takai M, Arakawa T, Tsumoto K. Control of protein adsorption to cyclo olefin polymer by the Hofmeister effect. J Pharm Sci. 2019. https://reader.elsevier.com/reader/sd/pii/S0022354919300012?token=2EE1009BB468D4D4F2336EDAE4258BB38D652A902A2D6905F5D1C7A5E40526F410BF48FC41203D89BAA43577AC8B9347 Accessed March 7, 2019.

  44. Pinholt C, Hartvig RA, Medlicott NJ, Jorgensen L. The importance of interfaces in protein drug delivery – why is protein adsorption of interest in pharmaceutical formulations? Expert Opin Drug Deliv. 2011;8(7):949–64.

    Article  CAS  PubMed  Google Scholar 

  45. Chang JY, Xiao NJ, Zhu M, et al. Leachables from saline-contained IV bags can alter therapeutic protein properties. Pharm Res. 2010;27:2402–13.

    Article  CAS  PubMed  Google Scholar 

  46. Mcleod AG, Walker IR, Zheng S, Hayward CPM. Loss of factor VIII activity during storage in PVC containers due to adsorption. Haemophilia. 2000;6(2):89–92.

    Article  CAS  PubMed  Google Scholar 

  47. Sreedhara A, Glover ZK, Piros N, Xiao N, Patel A, Kabakoff B. Stability of IgG1 monoclonal antibodies in intravenous infusion bags under clinical in-use conditions. J Pharm Sci. 2011;101(1):21–30.

    Article  CAS  PubMed  Google Scholar 

  48. Kumar O, Liu J, Ji J, Cheng W, Wang J, Joshi S, Middaugh CR, Volkin D. Compatibility, physical stability and characterization of an IgG4 monoclonal antibody after dilution into different intravenous administration bags. J Pharm Sci. 2012;101(10):3636–50.

    Article  CAS  Google Scholar 

  49. Mahler HC, Senner F, Maeder K, Mueller R. Surface activity of a monoclonal antibody. J Pharm Sci. 2009;98(12):4525–33.

    Article  CAS  PubMed  Google Scholar 

  50. Morar-Mitrica S, Puri M, Beumer Sassi A, Fuller J, Hu P, Crotts G, Nesta D. Development of a stable low-dose aglycosylated antibody formulation to minimize protein loss during intravenous administration. MAbs. 2015;7(4):792–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shi S, Hashemi V, Wang SC, Yang J, Yang MM, Semple A, Narasimhan C, Antochshuk V. Overcoming challenges with intravenous administration of an investigational protein therapeutic. J Pharm Sci. 2017;106(12):3465–73.

    Article  CAS  PubMed  Google Scholar 

  52. Werner BP, Winter G. Particle contamination of parenteralia and in-line filtration of proteinaceous drugs. Int J Pharm. 2015;496(2):250–67.

    Article  CAS  PubMed  Google Scholar 

  53. Besheer A. Protein adsorption to in-line filters of intravenous administration sets. J Pharm Sci. 2017;106(10):2959–65.

    Article  CAS  PubMed  Google Scholar 

  54. Pardeshi NN, Qi W, Dahi K, Caplan L, Carpenter JF. Microparticles and nanoparticles delivered in intravenous saline and in an intravenous solution of a therapeutic antibody product. J Pharm Sci. 2017;106(2):511–20.

    Article  CAS  PubMed  Google Scholar 

  55. Liu L, Randolph TW, Carpenter JF. Particles shed from syringe filters and their effects on agitation-induced protein aggregation. J Pharm Sci. 2012;101(8):2952–9.

    Article  CAS  PubMed  Google Scholar 

  56. Product information for trastuzumab emtansine (Kadcyla). https://www.tga.gov.au/sites/default/files/auspar-trastuzumab-emtansine-140317-pi.pdf. Accessed March 7, 2019.

  57. Werner BP, Winter G. Expanding bedside Filtrationd-a powerful tool to protect patients from protein aggregates. Usefulness of the final filter of the IV infusion set in intravenous administration of drugs-contamination. J Pharm Sci. 2018;107:2775–88.

    Article  CAS  PubMed  Google Scholar 

  58. Kuramoto K, Shoji T, Nakagawa Y. Usefulness of the final filter of the IV infusion set in intravenous administration of drugs-contamination of injection preparations by insoluble microparticles and its causes. Yakugaku Zasshi. 2006;126:289–95.

    Article  CAS  PubMed  Google Scholar 

  59. Ruzickova K, Cobbing M, Rossi M, Belazzi T. Preventing harm from phthalates, avoiding PVC in hospitals. http://www.accessmedicalsupply.com/content/preventing_harm_from_phthalates_avoiding_pvc_in_hospitals.pdf. Accessed March 7, 2019.

  60. Food and Drug Administration. Safety assessment of di(2-ethylhexyl) phthalate (DEHP) released from PVC medical devices. 2010. https://www.eldonjames.com/wp-content/uploads/2017/08/FDA_DEHP_PVC.pdf. Accessed March 6, 2019.

  61. Cheung JK, Sharma M, Dabbara A, Petersen J. The effect of formulation excipients on leachables for iv-administered products. 2012. https://www.pharmoutsourcing.com/Featured-Articles/125938-The-Effect-of-Formulation-Excipients-on-Leachables-for-IV-Administered-Products Accessed March 7, 2019.

  62. FDA warns against using Treanda Injection (solution) with closed system transfer devices, adapters, and syringes containing polycarbonate or acrylonitrile-butadiene-styrene; provides list of compatible devices. https://www.fda.gov/Drugs/DrugSafety/ucm437469.htm. Accessed Feb 12, 2019.

  63. Kiese S, Papppenberger A, Friess W, Mahler HC. Shaken, not stirred: mechanical stress testing of an IgG1 antibody. J Pharm Sci. 2008;97(10):4347–66.

    Article  CAS  PubMed  Google Scholar 

  64. Koepf E, Eisele S, Schroeder R, Brezesinski G, Friess W. Notorious but not understood: how liquid-air interfacial stress triggers protein aggregation. Int J Pharm. 2018;537(1-2):202–12.

    Article  CAS  PubMed  Google Scholar 

  65. Rudiuk S, Cohen-Tannoudji L, Huille S, Tribet C. Importance of the dynamics of adsorption and of a transient interfacial stress on the formation of aggregates of IgG antibodies. Soft Matter. 2012;8(9):2651–61.

    Article  CAS  Google Scholar 

  66. Bee JS, Schwartz DK, Trabelsi S, Freund E, Stevenson J, Carpenter JF, Randolph TW. Production of particles of therapeutic proteins at the air–water interface during compression/dilation cycles. Soft Matter. 2012;8(40):10329–35.

    Article  CAS  Google Scholar 

  67. Shieh I, Patel AR. Predicting the agitation-induced aggregation of monoclonal antibodies using surface tensiometry. Mol Pharm. 2015;12:3184–93.

    Article  CAS  PubMed  Google Scholar 

  68. Lee HJ, McAuley A, Schilke KF, McGuire J. Molecular origins of surfactant-mediated stabilization of protein drugs. Adv Drug Deliv Rev. 2011;63(13):1160–71.

    Article  CAS  PubMed  Google Scholar 

  69. Patapoff T, Esue O. Polysorbate 20 prevents the precipitation of a monoclonal antibody during shear. Pharm Dev Technol. 2009;14(6):659–64.

    Article  CAS  PubMed  Google Scholar 

  70. Singh SM, Bandi S, Jones DNM, Mallela KMG. Effect of polysorbate 20 and polysorbate 80 on the higher-order structure of a monoclonal antibody and its fab and fc fragments probed using 2d nuclear magnetic resonance spectroscopy. J Pharm Sci. 2017;106(12):3486–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Agarkhed M, O’Dell C, Hsieh MC, Zhang J, Goldstein J, Srivastava A. Effect of surfactants on mechanical, thermal, and photostability of a monoclonal antibody. AAPS PharmSciTech. 2018;19(1):79–92.

    Article  CAS  PubMed  Google Scholar 

  72. Wang S, Wu G, Zhang X, Tian Z, Hu T, Dai W, Qian F. Stabilizing two IgG1 monoclonal antibodies by surfactants: balance between aggregation prevention and structure perturbation. Eur J Pharm Biopharm. 2017;114:263–77.

    Article  CAS  PubMed  Google Scholar 

  73. John WM. Microbiological quality of drug products after penetration of the container system for dose preparation prior to patient administration. 2009. https://www.americanpharmaceuticalreview.com/Featured-Articles/114491-Microbiological-Quality-of-Drug-Products-after-Penetration-of-the-Container-System-for-Dose-Preparation-Prior-to-Patient-Administration/. Accessed March 7, 2019.

  74. Wu HC, Chen TN, Kao SH, Shui HA, Chen WJ, Lin HJ, Chen HM. Isoelectric focusing management: an investigation for salt interference and an algorithm for optimization. J Proteome Res. 2010;9(11):5542–56.

    Article  CAS  PubMed  Google Scholar 

  75. Haybrard J, Simon N, Danel C, Pinçon C, Barthélémy C, Tessier FJ, Décaudin B, Boulanger E, Odou P. Factors generating glucose degradation products in sterile glucose solutions for infusion: statistical relevance determination of their impacts. Sci Rep. 2017;7:11932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. USP. Dextrose injection. Pharmacopeial Forum. 2004;30(5):1614.

    Google Scholar 

  77. Glover ZW, Gennaro L, Yadav S, Demeule B, Wong PY, Sreedhara A. Compatibility and stability of pertuzumab and trastuzumab admixtures in i.v. infusion bags for coadministration. J Pharm Sci. 2013;102(3):794–812.

    Article  CAS  PubMed  Google Scholar 

  78. Narhi LO, et al. Subvisible (2–100 μm) particle analysis during biotherapeutic drug product development: Part 1, considerations and strategy. J Pharm Sci. 2015;104:1899–908.

    Article  CAS  PubMed  Google Scholar 

  79. Vincent C, et al. Subvisible (2–100 μm) particle analysis during biotherapeutic drug product development: Part 2, experience with the application of subvisible particle analysis. Biologicals. 2015;43(6):457–73.

    Article  CAS  Google Scholar 

  80. Bardin C, Astier A, Vulto A, Sewell G, Vigneron J, Trittler R, Daouphars M, Paul M, Trojniak M, Pinguet F. Guidelines for the practical stability studies of anticancer drugs: a European consensus conference. Ann Pharm Fr. 2011;69:221–31.

    Article  CAS  PubMed  Google Scholar 

  81. Khandpour RS. Handbook of biomedical instrumentation. New York: McGraw Hill; 2014. Chapter 68, Parenteral infusion devices.

    Google Scholar 

  82. Robert AP, James HP. Critical parameters in drug delivery by intravenous infusion. Expert Opin Drug Deliv. 2013;10(8):1095–108.

    Article  CAS  Google Scholar 

  83. U.S. Food & Drug Administration. What Is an Infusion Pump? 2017. https://www.fda.gov/MedicalDevices/ProductsandMedicalProcedures/GeneralHospitalDevicesandSupplies/InfusionPumps/ucm202495.htm. Accessed March 7, 2019.

  84. Thoele K, Piddoubny M, Ednalino R, Terry CL. Optimizing drug delivery of small-volume infusions. J Infus Nurs. 2018;41(2):113–7.

    Article  PubMed  Google Scholar 

  85. USP <787> Subvisible particulate matter in therapeutic protein injections. USP 41: 6534-6536.

    Google Scholar 

  86. Galush WJ, Horst TA. Vented spikes improve delivery from intravenous bags with no air headspace. J Pharm Sci. 2015;104:2397–400.

    Article  CAS  PubMed  Google Scholar 

  87. Naoki Asakawa, The horror of sample adsorption to containers, Shimadzu Corporation, https://www.shimadzu.com/an/hplc/support/lib/lctalk/96/96uhplc.html. Accessed September 22, 2018.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Supriya Gupta .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2020 American Association of Pharmaceutical Scientists

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Gupta, S. et al. (2020). Chapter 18: Design of Clinical In-Use Studies. In: Jameel, F., Skoug, J., Nesbitt, R. (eds) Development of Biopharmaceutical Drug-Device Products. AAPS Advances in the Pharmaceutical Sciences Series, vol 35. Springer, Cham. https://doi.org/10.1007/978-3-030-31415-6_18

Download citation

Publish with us

Policies and ethics