Developing the Subcutaneous Drug Delivery Route

Main Article Content

J. M. Wright Graham B. Jones

Abstract

Biopharmaceuticals and biosimilars now represent the majority of the top 10 selling drugs. High development costs and requirement for most to be administered intravenously are noted drawbacks. Recent advances in subcutaneous drug delivery methods offer a viable alternative route for parenteral drug administration. Comparison studies have highlighted equivalent drug efficacies coupled with substantial cost savings. Allied to these developments are new closed-loop drug delivery systems which have the potential to revolutionize the biopharmaceutical sector through active patient engagement. First demonstrated in the insulin marketplace, these efforts represent formal embodiments of the precision medicine approach to disease management. This review will highlight key considerations for subcutaneous drug delivery, including patient preferences, drug formulation and needle and device design. We also provide an overview of the market evolution of subcutaneously administered drugs, highlighting those currently in clinical development, and predict areas for future innovation.

Keywords: Subcutaneous, closed loop, interstitial matrix, hyaluronidase, biologics, drug development, personalized medicine, syringe and needle technology

Article Details

How to Cite
WRIGHT, J. M.; JONES, Graham B.. Developing the Subcutaneous Drug Delivery Route. Medical Research Archives, [S.l.], v. 5, n. 12, dec. 2017. ISSN 2375-1924. Available at: <https://esmed.org/MRA/mra/article/view/1652>. Date accessed: 29 mar. 2024. doi: https://doi.org/10.18103/mra.v5i12.1652.
Section
Research Articles

References

References
1. Mullard A. Robust Biotech Sector Increases R&D Spend. Nature Reviews Drug Discovery 14, 449 (2015).
2. Collins FS, Varmus H. A New Initiative on Precision Medicine. N. Engl. J. Med. 372, 793-795 (2015).
3. McCabe C, Claxton, K, Culyer AJ. The NICE Cost-Effectiveness Threshold What it is and What that Means, Pharma-coeconomics 26, 733-744 (2008).
4. Brinckerhoff CC, Schorr K. Patent Watch: Have The Biosimilar Floodgates Been Opened in the United States? Nature Reviews Drug Discovery 14, 303–304 (2015).
5. Tetteh EK, Morris S. Evaluating the administration costs of biologic drugs: development of a cost algorithm. Health Economics Review 4, 26 (2014).
6. Jin J, Zhu LL, Chen M, Xu HM, Wang HF, Feng XQ, Zhou XP. The Optimal Choice of Medication Administration Route Regarding Intravenous, Intramuscular, and Subcutaneous Injection. Patient Preference and Adherence 9, 923–942 (2015).
7. Rejinold NS, Shin JH, Seok HY, Kim YC. Biomedical Applications of Microneedles in Therapeutics: Recent Advancements and Implications in Drug Delivery. Expert Opinion on Drug Delivery 13, 109-131 (2016).
8. Trevaskis NL, Kaminskas LM, Porter CJ. From Sewer to Saviour -Targeting the Lymphatic System to Promote Drug Exposure and Activity. Nat. Rev. Drug. Disc. 14, 781 (2015).
9. Berteau C, Filipe-Santos O, Wang T, Rojas HE, Granger C, Schwarzenbach F. Evaluation of the Impact of Viscosity, Injection Volume and Injection Flow Rate on Subcutaneous Injection Tolerance. Medical Devices:Evidence and Research 8 473–484 (2015).
10. Harvey AJ, Kaestner SA, Sutter DE, Harvey NG, Mikszta JA, Pettis RJ.
Microneedle-Based Intradermal Delivery Enables Rapid Lymphatic Uptake and Distribution of Protein Drugs. Pharm. Res. 28, 107-116 (2011).
11: Wright JM, Thyagarajapuram NR, Sirkar R, Kapur S, Harrison MW, Collins DS, Bryan DJ, Jones GB. Optimizing the bioavailability of subcutaneously administered therapeutics through mechanochemical drivers Pharm. Res, 2017, 34(10), 2000-2011.
12: Sawinski VJ, Loiselle RJ, Goldberg AF. Histologic evaluation of injection site edema. Oral Surg Oral Med Oral Pathol. 1966, 22(1):125-6.
13: Kurtin S, Knop CS, Milliron T. Subcutaneous Administration of Bortezomib: Strategies to Reduce Injection Site Reactions, J Adv Pract Oncol. 2012, 3(6): 406–410.
14. Shapiro R. Subcutaneous Immunoglobulin Therapy by Rapid Push is Preferred to Infusion by Pump: A Retrospective Analysis. J. Clin. Immunol. 30, 301-30 (2010).
15. Yang MX, Shenoy B, Disttler M, Patel R, McGrath M, Pechenov S, Margolin AL. Crystalline Monoclonal Antibodies for Subcutaneous Delivery. Proc. Natl. Acad. Sci. USA, 100, 6934–6939 (2003).
16. Schoellhammer CM, Schroeder A, Maa R, Lauwers GY, Swiston A, Zervas M, Barman R, DiCiccio AM, Brugge WR, Anderson DG, Blankschtein D, Langer R, Traverso G. Ultrasound-Mediated Gastrointestinal Drug Delivery. Sci. Transl. Med. 7 310ra168 (2015).
17. Fathallah AM, Turner MR, Mager DE, Balu-Iyer SV. Effects of Hypertonic Buffer Composition on Lymph Node Uptake and Bioavailability of
Rituximab, After Subcutaneous Administration. Biopharm Drug Dispos. 36, 115–125 (2015).
18. Bocci V, Muscettola M, Grasso G, Magyar Z, Naldini A, Szabo G.The lymphatic route. 1) Albumin and hyaluronidase modify the normal distribution of interferon in lymph and plasma. Experientia, 42 (4) 432–433 (1986).
19. Fathallah AM, Balu-Iyer SV. Anatomical, Physiological, and Experimental Factors Affecting the Bioavailability of sc-Administered Large Biotherapeutics. J. Pharm. Sci. 104, 301–306 (2015). Deng R, Meng YG, Hoyte K, Lutman J, Lu Y, SuhasiniIyer I, DeForge LE, Theil F-P, Fielder PJ, Prabhu S. Subcutaneous Bioavailability of Therapeutic Antibodies as a Function of FcRn Binding Affinity in Mice, mAbs 4, 101-109 (2012).
20. Richter WF, Jacobsen B. Subcutaneous Absorption of Biotherapeutics: Knowns and Unknowns. Drug Metab Dispos. 42, 1881-1889 (2014).
21. Kang DW, Jadin LM, Nekoroski TA, Zepeda ML. Recombinant Human Hyaluronidase (rHuPH20) Facilitates Subcutaneous Infusion of Immunoglobulin, Increases Local Fluid Dispersion, and Reduces Induration in a Porcine Model. J. Allerg. Clin.Immunol. 129(2) AB85 (2012).
22. Kang DW, Jadin L, Nekoroski T, Drake FH, Zepeda ML. Recombinant Human Hyaluronidase PH20 (rHuPH20) Facilitates Subcutaneous Infusions of Large Volumes of Immunoglobulin in a Swine Model. Drug Deliv. Transl. Res 2, 254-264 (2012); Bookbinder LH et al. A recombinant human enzyme for enhanced interstitial transport of therapeutics. J Control Release 114(2), 230-41 (2006).
23. Zheng Y. et al. Minipig as a Potential Translatable Model for Monoclonal Antibody Pharmacokinetics After Intravenous and Subcutaneous Administration. mAbs 4, 243-255 (2012).
24. Ismael G, et al. Subcutaneous Versus Intravenous Administration of (neo) Adjuvant Trastuzumab in Patients with HER2-Positive, Clinical Stage I–III Breast Cancer (HannaH study): A Phase 3, Open-Label, Multicentre, Randomised Trial. Lancet Oncol. 13, 869–78 (2012); Pivot X. et al. Preference for Subcutaneous or Intravenous Administration of Trastuzumab in Patients with HER2-Positive Early breast cancer (PrefHer): An Open-Label Randomised Study. Lancet Oncol. 14(10), 962-970 (2013).
25. Salar A. et al. Comparison of Subcutaneous Versus Intravenous Administration of Rituximab As Maintenance Treatment for Follicular Lymphoma: Results From a Two-Stage, Phase IB Study. J Clin Oncol. 32, 1782-1791 (2014).
26. Spandorfer PR. et al. A Randomized Clinical Trial of Recombinant Human Hyaluronidase-Facilitated Subcutaneous Versus Intravenous Rehydration in Mild to Moderately Dehydrated Children in the Emergency Department, Clinical Therapeutics, 34 (11), 2232-2245 (2012).
27: Jain M, Doughty D, Clawson C, Li X, White N, Agoram B, van der Merwe R. Tralokinumab pharmacokinetics and tolerability when administered by different subcutaneous injection methods and rates, International Journal of Clinical Pharmacology and Therapeutics, 2017, 55(7):606-618.
28. Comley K, Fleck NA. Deep penetration and liquid injection into adipose tissue, J. Mech. Mater. Struct. 6, 127-140 (2011).
29. Thomsen M, Hernandez-Garcia A, Mathiesen J, Poulsen M, Sørensen DN, Tarnow L, Feidenhans R. Model Study of the Pressure Build-Up during Subcutaneous Injection. PLOS One 9(8) e104054 (2014).
30: Kim H, Park H, Lee SJ. Effective method for drug injection into subcutaneous tissue, Scientific Reports 2017, 7, # 9613 doi:10.1038/s41598-017-10110-w
31. Videira MA, Botelho MF, Santos AC, Gouveia LF, Pedroso de Lima JJ, Almeida AJ. Lymphatic Uptake of Pulmonary Delivered Radiolabelled Solid Lipid Nanoparticles. J. Drug Target. 10 (8), 607–613 (2002).
32. Zhang F, Niu G, Lu G, Chen X. Preclinical Lymphatic Imaging. Mol Imaging Biol. 13(4) 599–612 (2011).
33. Eberlin LS. et al. Visualizing Dermal Permeation of Sodium Channel Modulators by Mass Spectrometric Imaging. J. Am. Chem. Soc. 136, 6401-6405 (2014).
34. Doyle III FJ, Huyett LM, Lee JB, Zisser HC, Dassau E. Closed-Loop Artificial Pancreas Systems: Engineering the Algorithms. Diabetes Care. 37 (5), 1191-1197 (2014).
35. Bergenstal RM, Garg S, Weinzimer SA, Buckingham BA, Bode BW, Tamborlane WV, Kaufman FR. Safety of a Hybrid Closed-Loop Insulin Delivery System in Patients With Type 1 Diabetes, JAMA. 2016, 316 (13),1407-1408.
36: Mage PL, Ferguson BS, Maliniak D, Ploense KL, Kippin TE, Soh HT. Closed-loop control of circulating drug levels in live animals, Nature Biomedical Engineering 2017, 1, # 0070 doi:10.1038/s41551-017-0070.
37: Ilyas M, Butt MFU, Bilal M, Mahmood K, Khaqan A, Riaz RA. A Review of Modern Control Strategies for Clinical Evaluation of Propofol Anesthesia Administration Employing Hypnosis Level Regulation, BioMed Research International, 2017, https://doi.org/10.1155/ 017/7432310.
38. Farandos NM, 
Yetisen AK, 
 Monteiro MJ, Lowe CR,
Hyun Yun S. Contact Lens Sensors in Ocular Diagnostics, Advanced Healthcare Materials, 4(6), 785–937(2015).
39. Zhukov I, Mikhaylov D, Starikovskiy A. Nano sensors integrated into dental implants for detection of acute myocardial infarction, Int. J. Emerging Trends & Tech. in Comp. Sci, 1(2), 85-87 (2012).
40. Cash KJ, Clark HA. Nanosensors and nanomaterials for monitoring glucose in diabetes, Trends Mol Med. 16(12), 584–593 (2010).
41. Evaluation of performance, safety, subject acceptance, and compliance of a disposable autoinjector for subcutaneous injections in healthy volunteers, Berteau C, Schwarzenbach F, Donazzolo Y, Latreille M, Berube J, Abry H, Cotton J, Feger C, Laurent PE. Patient Preference and Adherence 4, 379–388 (2010).
42: Ward WK. A Review of the Foreign-body Response to Subcutaneously-implanted Devices: The Role of Macrophages and Cytokines in Biofouling and Fibrosis, J. Diabetes Sci. Technol. 2008, 2(5), 768-777.
43: Onuki Y, Bhardwaj U, Papadimitrakopoulos F, Burgess DJ. A Review of the Biocompatibility of Implantable Devices: Current Challenges to Overcome Foreign Body Response, J Diabetes Sci Technol 2008, 2(6):1003-1015.
44: Klopfleisch R, Jung F. The pathology of the foreign body reaction against biomaterials, J. Biomed. Mater. Res. Part A, 2017:105A:927–940.
45: Anderson JM, Rodriguez A, Chang DT. Foreign Body Reaction to Biomaterials, Response to Materials, Semin Immunol. 2008 Apr; 20(2): 86–100.
46: Wang Y, Vaddiraju S, Gu B, Papadimitrakopoulos F, Burgess DJ. Foreign Body Reaction to Implantable Biosensors: Effects of Tissue Trauma and Implant Size, J Diabetes Sci Technol. 2015 Sep; 9(5): 966–977.
47: Morais JM, Papadimitrakopoulos F, Burgess DJ. Biomaterials/Tissue Interactions: Possible Solutions to Overcome Foreign Body Response, AAPS J. 2010 Jun; 12(2): 188–196.
48: Norton LW, Koschwanez HE, Wisniewski NA, Klitzman B. Reichert, WM. Vascular endothelial growth factor and dexamethasone release from non fouling sensor coatings affect the foreign body response, J Biomed Mater Res A. 2007 June 15; 81(4): 858–869.
49: Sinclair KD, et al. Development of a Broad Spectrum Polymer Released Antimicrobial Coating for the Prevention of Resistant Strain Bacterial Infections, J. Biomed. Mater. Res. A. 2012, 100(10): 2732–2738.
50: Wang Y, Papadimitrakopoulos F, Burgess DJ. Polymeric "smart" coatings to prevent foreign body response to implantable biosensors, J Control Release. 2013 169(3):341-7.
51: Wang Y, Vaddiraju S, Qiang L, Xu X, Papadimitrakopoulos F, Burgess DJ. Effect of Dexamethasone - Loaded Poly(Lactic-Co-Glycolic Acid) Microsphere/Poly (Vinyl Alcohol) Hydrogel Composite Coatings on the Basic Characteristics of Implantable Glucose Sensors, J Diabetes Sci Technol. 2012 6: 1445–1453.
52: Lin P, Lin CW, Mansour R, Gu F. Improving biocompatibility by surface modification techniques on implantable bioelectronics, Biosens Bioelectron. 2013 Sep 15;47:451-60.
53: Bridges AW, García AJ. Anti-Inflammatory Polymeric Coatings for Implantable Biomaterials and Devices, Journal of Diabetes Science and Technology Volume 2, Issue 6, 2008, 984-994.
54: Bhardwaj U, Sura R, Papadimitrakopoulos F, Burgess DJ. PLGA/PVA hydrogel composites for long-term inflammation control following s.c. implantation; Int J Pharm. 2010 Jan 15;384(1-2):78-86.
55: Zhang L, Cao Z, Bai T, Carr L, Ella-Menye J-R, Irvin C, Ratner BD, Jiang S. Zwitterionic hydrogels implanted in mice resist the foreign-body reaction. Nature Biotechnology 2013, 31 (6), 553-557.
56: Patel PJ. et al. Randomized Trial of Infusion Set Function: Steel Versus Teflon, Diabetes Tech. Therapeutics, 2014, 16, 15-19.
57: Avula MN. et al. Local release of masitinib alters in vivo implantable continuous glucose sensor performance; Biosens Bioelectron. 2016 Mar 15;77:149-56.
58: Avula MN, Rao AN, McGill LD, Grainger DW, Solzbacher F. Modulation of the foreign body response to implanted sensor models through device-based delivery of the tyrosine kinase inhibitor, masitinib, Biomaterials. 2013 34(38):9737-46.
59: Patil SD, Papadmitrakopoulos F, Burgess DJ. Concurrent delivery of dexamethasone and VEGF for localized inflammation control and angiogenesis, J. Contr. Rel. 2007, 117, 68–79.
60: Ward WK, Hansen JC, Massoud RG, Engle JM, Takeno MM, Hauch KD. Controlled release of dexamethasone from subcutaneously-implanted biosensors in pigs: localized anti-inflammatory benefit without systemic effects, J Biomed Mater Res A. 2010 Jul;94(1):280-7.
61: Bis RL, Mallela KMG. Antimicrobial preservatives induce aggregation of interferon alpha-2a: The order in which preservatives induce protein aggregation is independent of the protein, Int. J. Pharm. 2014, 472(0): 356–361.
62: Kim D, Baraniuk J. Delayed-type hypersensitivity reaction to the meta cresol component of insulin, Ann. Allergy Asthma Immunol., 2007, 99, 194-195.
63: Paiva TO, Bastos AEP, Marques JT, Viana AS, Limab PA, deAlmeida RFM. m-Cresol affects the lipid bilayer in membrane models and living neurons, RSC Adv. 2016, 6, 105699–105712.
64: Lima DP, Diniz DG, Moimaz SAS, Sumida DH, Okamoto AC. Saliva: reflection of the body, International Journal of Infectious Diseases, 2010, 14, e184-188.
65: Le Guezennec X, Quah J, Tong L, Kim N. Human tear analysis with miniaturized multiplex cytokine assay on “wall-less” 96-well plate, Mol Vis. 2015, 21, 1151–1161.
66: Robb DM, Kanji Z. Comparison of two needle sizes for subcutaneous administration of enoxaparin: effects on size of hematomas and pain on injection, Pharmacotherapy. 2002, 22(9):1105-9.
67: Hirsch L, Gibney M, Berube J, Manocchio J. Impact of a Modifed Needle Tip Geometry on Penetration Force as well as Acceptability, Preference, and Perceived Pain in Subjects with Diabetes, Journal of Diabetes Science and Technology, 2012, 6(2), 328-335.
68: Guerreiro-Tanomaru JM, Loiola LE, Morgental RD, Leonardo R, Tanomaru-Filho M. Efficacy of four irrigation needles in cleaning the apical third of root canals, Braz. Dent. J. 2013, 24(1), http://dx.doi.org/10.1590/0103-6440201302153.
69: Edsberg B, Herly D, Hildebrandt P, Kuhl C. Insulin bolus given by sprinkler needle: effect on absorption and glycaemic response to a meal, British Medical Journal, 1987, 294, 1373-1376.
70: Jones GB, Collins DS, Harrison MW, Thayagarjapuram NR, Wright JM. Personalizing Drug Delivery: Exploiting the Subcutaneous Revolution, Science Translational Medicine, 2017, 9 (405), eaaf9166.
71. Stoner, KL, Harder H, Fallowfield LJ, Jenkins VA. Intravenous versus Subcutaneous Drug Administration. Which Do Patients Prefer? A Systematic Review. The Patient, 8, 145-153 (2015).
72. Dias C, Abosaleem B, Crispino C, Gao B, Shaywitz A. Tolerability of High-Volume Subcutaneous Injections of a Viscous Placebo Buffer: A Randomized, Crossover Study in Healthy Subjects. AAPS PharmSciTech. 16, 1101-1107 (2015).
73. Masid MLS. et al. A Patient Care Program for Adjusting the Autoinjector Needle Depth According to Subcutaneous Tissue Thickness in Patients With Multiple Sclerosis Receiving Subcutaneous Injections of Glatiramer Acetate. J. Neuroscience Nursing 47 (1) E23 (2015).
74: Asplund R. Manual lymph drainage therapy using light massage for fibromyalgia sufferers: a pilot study. Journal of Orthopaedic Nursing. 2003; 7:192–196.
75: Resende MA, Sabino GG, Candido CRM, Pereira LSM, Francischi JN. Local transcutaneous electrical stimulation (TENS) effects in experimental inflammatory edema and pain, European Journal of Pharmacology, 2004, 504, 217–222.
76. Carmichael JD. Lanreotide depot deep subcutaneous injection: a new method of delivery and its associated benefits. Patient Preference and Adherence 6, 73–82 (2012);
77. Jackisch C, Müller V, Maintz C, Hell S, Ataseven B. Subcutaneous Administration of Monoclonal Antibodies in Oncology. Geburtshilfe Frauenheilkd 74(4), 343–349 (2014);
78. Wynne CJ. et al. Comparative pharmacokinetics of subcutaneous trastuzumab administered via handheld syringe or proprietary single-use injection device in healthy males. Cancer Chemotherapy and Pharmacology 72 (5), 1079-1087 (2013).
79. Martin JR, Beegle NL, Zhu Y, Hanisch EM. Subcutaneous Administration of Bortezomib: A Pilot Survey of Oncology Nurses. J. Adv. Pract. Oncol. 6, 308-318 (2015).
80. Samanta K, Moore L, Jones G, Evason J, Owen G. PCN39 Potential Time and Cost Savings with Herceptin (Trastuzumab) Subcutaneous (SC) Injection Versus Herceptin Intravenous (IV) Infusion: Results from Three Different English Patient Settings. Value Health 15 (7), A415 (2012).
81. Amur S, LaVange L, Zineh I, Buckman-Garner S, Woodcock J. Biomarker Qualification: Toward a Multiple Stakeholder Framework for Biomarker Development, Regulatory Acceptance, and Utilization Clin. Pharmacol. Ther. 2015, 98, 34–46.
82. FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource. Silver Spring (MD): Food and Drug Administration (US); 2016-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK326791/
83. In Vitro Companion Diagnostic Devices: Guidance for Industry and Food and Drug Administration Staff, 8/6/14:
https://www.fda.gov/downloads/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/UCM262327.pdf
84. FDA 2016: Principles for Codevelopment of an In Vitro Companion Diagnostic Device with a Therapeutic Product : Draft Guidance for Industry and Food and Drug Administration Staff, 7/15/16:https://www.fda.gov/downloads/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/UCM510824.pdf
85. Blackstone EA, Joseph PF. The Economics of Biosimilars, Am Health Drug Benefits. 2013, 6(8), 469–478.
86. Knibb R, Morton K. Accuracy in use of adrenalin auto-injectors in a simulated emergency situation: a comparison of JEXT, EpiPen and Emerade, Clin Transl Allergy. 2015, 5(S3), O5.
87.Schwirtz A, Seeger H. Comparison of the robustness and functionality of three adrenaline auto-injectors, J Asthma Allergy. 2012, 5, 39–49.
88. Guerlain S, Hugine A, Wang L. A comparison of 4 epinephrine autoinjector delivery systems: usability and patient preference, Ann Allergy Asthma Immunol. 2010, 104(2), 172–177.
89. Ventola CL. Biosimilars Part 1: Proposed Regulatory Criteria for FDA Approval, Pharmacy and Therapeutics, 2013, 38(5): 270-274.
90. Berkowitz SA, Engen JR, Mazzeo JR, Jones GB. Analytical tools for characterizing biopharmaceuticals and the implications for biosimilars. Nature Reviews Drug Discovery, 2012, 11, 527-540.