The Subcutaneous Route of Administration for Therapeutic Antibodies: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 1 by Aubin Pitiot.

Therapeutic antibodies (Abs) are used in the treatment of numerous diseases, including infection, cancer, and autoimmune disorders, in which they have already demonstrated their efficacy. The success of Abs is due to (I) a high level of specificity and affinity to their target antigen, (II) a favorable safety profile, and (III) a unique pharmacokinetic profile, supporting a longer half-life as compared to other drugs. While the majority of Abs are delivered through IV injection, evidence in the literature showed that beyond the reduction of invasiveness, a better efficacy could be achieved with a local delivery of Abs. Among those routes, one has been in full development these last few years, with already good clinical results and still promising developments : The Subcutaneous route.

  • Subcutaneous delivery
  • Drug Administration
  • Therapeutic Antibodies
  • Clinical Trials

1. Introduction

Over the past 30 years, therapeutic antibodies (Abs) have been found to be valuable therapeutics [1]. A total of 6 to 12 new Abs are approved by the U.S. FDA and/or the EMA each year, and new molecules are reaching clinical trials every month [2]. Therapeutic antibodies are used in the treatment of numerous diseases, including infection, cancer, and autoimmune disorders, in which they have already demonstrated their efficacy [3][4].
The success of Abs is due to (I) a high level of specificity and affinity to their target antigen, (II) a favorable safety profile, and (III) a unique pharmacokinetic profile, supporting a longer half-life as compared to other drugs [5]. These characteristics have allowed Abs to move rapidly from pre-clinical studies to clinical trials, as observed during the COVID-19 pandemic [6].
From the historical full-length antibody, molecular engineering has enabled the development of multiple and diverse Ab formats, including multi-specific Abs, fragments, and conjugated Abs that are now extensively evaluated in clinical trials [7].
Due to their intrinsic biological properties, Abs have a specific interconnected pharmacokinetic and pharmacodynamic profile, which influence their absorption and biodistribution after administration [5]. Abs pharmacokinetics is linked to their route of administration [8]. Historically, Abs were delivered via intravenous (IV) injection. Nowadays, the subcutaneous (SC) route is often used for chronic diseases [9]. These systemic routes have the advantage of allowing the delivery of large amounts of Abs and to enable rapid systemic bioavailability. However, one of their drawbacks is the limited distribution from the site of injection via the blood flow to the diseased organ, which may result in limited Ab amount in the vicinity of the target antigen. Ultimately, this necessitates the injection of a high dose, which may be associated with potential toxicity and cost issues. Accumulating preclinical evidence has driven researchers to reconsider Abs’ route of administration in order to maximize their therapeutic index.
Alternative delivery methods, addressing Abs to the disease site (e.g., delivery of Abs in the lung to treat respiratory pathologies [10], or inside a tumor [11]) have emerged and progressed to the clinical trial stage. In theory, a higher concentration of the antibody at the target site should improve the therapeutic response, while lowering the concentration in neighboring healthy tissues, resulting in reduced side effects. One of those favorable route of administration, discussed here, is the subcutaneous injection.

2. The Subcutaneous Route: The Most Popular after IV Injection

2.1. Fundamentals Related to the SC Route

After IV injection, the second most popular route for the delivery of antibodies is the subcutaneous (SC) route. It consists in the injection of Abs using a syringe and needle under the skin of patients at an angle of 90 °C, thus bypassing the barrier formed by the epidermis and dermis layers [12]. The choice of the anatomical site is important due to differences in dermal thickness which may reduce the absorption of the injected Abs. Nowadays, around 30% of the approved Abs are delivered by SC injection (Table 1). If the delivery of drugs, mainly opioids, by SC administration, has been in practice since the middle of the 19th century, the administration of Abs by this route is recent. The first subcutaneous injected Ab was Adalimumab, used in the treatment of rheumatoid arthritis and approved by the FDA in 2002, and by the EMA in 2003 [13]. After this first success, and particularly since 2009, the number of marketed Abs delivered by the SC route has significantly increased. It is noteworthy that SC administration is already the standard route in the treatment of chronic diseases such as rheumatoid arthritis. Indeed, it allows self-administration and improves patients’ compliance. The SC route is mainly used for the delivery of Abs targeting interleukins such as TNF-α, critically involved in the development of rheumatoid arthritis (Adalimumab, Golimumab, Certolizumab pegol) or cytokine receptors such as the IL-17a receptor, involved in the progression of psoriasis (Brodalumab, Secukinumab, Ixekizumab) [14]. The development of Abs intended for a subcutaneous injection necessitates understanding the physiology of the skin. After injection, the drug reaches the hypodermis interstitial space between the dermis and the deep fascia covering the muscle tissue. This layer is composed of adipose tissue, blood, lymph vessels, and resident immune cells such as fibroblasts and macrophages. All components are enmeshed in an extracellular matrix (ECM) network, rich in collagen, elastin, and glycosaminoglycans [15]. To pass into the systemic compartment (via either the blood capillaries or lymphatic vessels), and thus reach their target, Abs have to diffuse through the ECM, which constitutes both a physical and chemical barrier. The fate of the Ab is dictated by its size, charge, and affinity with transporters. Despite the presence of the positively charged collagen fibrils, the hypodermis interstitial space displayed an overall negative charge due to important concentrations of hyaluronic acid and chondroitin sulfate, two major glycosaminoglycans of the hypodermis ECM, which are negatively charged. The global negative charge of ECM favors the transport of negatively charged drugs thanks to electrostatic repulsion [16]. However, the majority of therapeutic Abs are positively charged. Once the ECM is traversed, drugs may enter the systemic circulation by two different mechanisms. Molecules smaller than 16 kDa diffuse directly into the bloodstream, taking advantage of the permeability of the vascular endothelium [16]. However, Abs, along with drugs with a higher molecular weight, are absorbed by convection into lymphatic vessels. Thus, the subcutaneous route is particularly interesting to target lymphoid cells and the molecules they secrete. Abs in the lymphatic vessels pass to larger lymphatics and then reach the blood vascular system, from where they diffuse throughout the body. If the development of Abs for subcutaneous injection is quite challenging, multiple factors explain the attractiveness of this route as compared to other parenteral ones. In the hypodermis, the walls in the fat lobule are thinner than those in the dermis, which facilitates the diffusion of drugs into blood capillaries [17]. Moreover, the absence of antigen-presenting cells in the hypodermis, usually present in the top layers of the skin (Langerhans cells and/or macrophages), may decrease the immunogenicity of the antibody. Thus, an increasing number of Abs delivered by SC are being developed, allowing a quicker delivery time of administration as compared to IV injection, enabling longer dosing intervals and, in fine, reducing the frequency of administration. In addition, SC administration is less invasive and painful than IV injection [16] and allows self-delivery at home [18]. Thus, the subcutaneous route may improve patient comfort and compliance, which is critical for the treatment of chronic diseases, and may be associated with a reduction in treatment costs, consuming fewer healthcare resources.

2.2. Abs Approved for Subcutaneous Delivery

Abs approved for subcutaneous administration must be formulated at a high concentration, thus necessitating a careful control of their stability and formulation viscosity. Different strategies have been considered to ensure the efficient absorption and bioavailability of Abs after hypodermis injection. They include, but are not limited to, the increase in delivered Abs concentration (e.g., SC administration limiting the injection volume to 1–2 mL [19]), the development of specific formulations to reduce physical and chemical destabilization (e.g., the use of polysorbate preventing aggregation and particle formation [20]) and the development of novel administration devices (e.g., the autoinjectors enabling a faster delivery for larger concentrations of Abs [21]). Those strategies have led to the approval of around 40 different Abs (Table 1).
Table 1.
Therapeutic antibodies approved by the Food and Drug Administration (FDA) or the European Medicine Agency (EMA) for subcutaneous administration.

References

  1. Asher Mullard; FDA approves 100th monoclonal antibody product. Nature Reviews Drug Discovery 2021, 20, 491-495, 10.1038/d41573-021-00079-7.
  2. Antibody therapeutics approved or in regulatory review in the EU or US . Antibody Society. Retrieved 2022-9-8
  3. David Zahavi; Louis Weiner; Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34, 10.3390/antib9030034.
  4. Lawrence Steinman; The use of monoclonal antibodies for treatment of autoimmune disease. Journal of Clinical Immunology 1990, 10, 30S-39S, 10.1007/bf00918689.
  5. Josiah T. Ryman; Bernd Meibohm; Pharmacokinetics of Monoclonal Antibodies. CPT: Pharmacometrics & Systems Pharmacology 2017, 6, 576-588, 10.1002/psp4.12224.
  6. Lin Ning; Hamza B. Abagna; Qianhu Jiang; Siqi Liu; Jian Huang; Development and application of therapeutic antibodies against COVID-19. International Journal of Biological Sciences 2021, 17, 1486-1496, 10.7150/ijbs.59149.
  7. Mariam Elshiaty; Hannah Schindler; Petros Christopoulos; Principles and Current Clinical Landscape of Multispecific Antibodies against Cancer. International Journal of Molecular Sciences 2021, 22, 5632, 10.3390/ijms22115632.
  8. Alagga, A.A.; Gupta, V.. Drug Absorption; StatPearls Publishing: Treasure Island, FL, USA, 2022; pp. 1.
  9. Andrea Matucci; Alessandra Vultaggio; Romano Danesi; The use of intravenous versus subcutaneous monoclonal antibodies in the treatment of severe asthma: a review.. Respiratory Research 2018, 19, 154, 10.1186/s12931-018-0859-z.
  10. Elsa Bodier-Montagutelli; Renaud Respaud; Hervé Watier; Audrey Guillon-Munos; MAbDelivery: Administration routes for antibody therapy Third LabEx MAbImprove industrial workshop, July 2, 2015 Tours, France. Platform development for expression and purification of stable isotope labeled monoclonal antibodies in Escherichia coli 2017, 9, 579-585, 10.1080/19420862.2017.1298899.
  11. Stéphane Champiat; Lambros Tselikas; Siham Farhane; Thibault Raoult; Matthieu Texier; Emilie Lanoy; Christophe Massard; Caroline Robert; Samy Ammari; Thierry De Baère; et al.Aurélien Marabelle Intratumoral Immunotherapy: From Trial Design to Clinical Practice. Clinical Cancer Research 2021, 27, 665-679, 10.1158/1078-0432.ccr-20-0473.
  12. Hyejeong Kim; Hanwook Park; Sang Joon Lee; Effective method for drug injection into subcutaneous tissue. Scientific Reports 2017, 7, 1-11, 10.1038/s41598-017-10110-w.
  13. Furst, D.E.; Schiff, M.H.; Fleischmann, R.M.; Strand, V.; Birbara, C.A.; Compagnone, D.; Fischkoff, S.A.; Chartash, E.K; Adalimumab, a fully human anti tumor necrosis factor-alpha monoclonal antibody, and concomitant standard antirheumatic therapy for the treatment of rheumatoid arthritis: Results of STAR (Safety Trial of Adalimumab in Rheumatoid Arthritis). J. Rheumatol. 2003, 30, 2563-2571.
  14. Gupta, A.K.; Versteeg, S.V.; Abramovits, W.; Vincent, K.D.; Brodalumab (Siliq®): A Treatment for Plaque Psoriasis. SKINmed 2017, 15, 281-285.
  15. Oktay Arda; Nadir Göksügür; Yalçın Tüzün; Basic histological structure and functions of facial skin. Clinics in Dermatology 2014, 32, 3-13, 10.1016/j.clindermatol.2013.05.021.
  16. Margarida Viola; Joana Sequeira; Raquel Seiça; Francisco Veiga; João Serra; Ana C. Santos; António J. Ribeiro; Subcutaneous delivery of monoclonal antibodies: How do we get there?. Journal of Controlled Release 2018, 286, 301-314, 10.1016/j.jconrel.2018.08.001.
  17. Irwin M. Braverman; Agnes Keh-Yen; Ultrastructure of the Human Dermal Microcirculation. III. The Vessels in the Mid- and Lower Dermis and Subcutaneous Fat. Journal of Investigative Dermatology 1981, 77, 297-304, 10.1111/1523-1747.ep12482470.
  18. Manuel Sánchez-Félix; Matt Burke; Hunter H. Chen; Claire Patterson; Sachin Mittal; Predicting bioavailability of monoclonal antibodies after subcutaneous administration: Open innovation challenge. Advanced Drug Delivery Reviews 2020, 167, 66-77, 10.1016/j.addr.2020.05.009.
  19. Neeraj Kohli; Nidhi Jain; Melissa L Geddie; Maja Razlog; Lihui Xu; Alexey A Lugovskoy; A novel screening method to assess developability of antibody-like molecules. mAbs 2015, 7, 752-758, 10.1080/19420862.2015.1048410.
  20. Wei Wang; Satish Singh; David L. Zeng; Kevin King; Sandeep Nema; Antibody Structure, Instability, and Formulation. Journal of Pharmaceutical Sciences 2007, 96, 1-26, 10.1002/jps.20727.
  21. Optimizing Drug Delivery for Modern Biologics . Pharmaceutical Technology. Retrieved 2022-9-8
  22. Asher Mullard; 2020 FDA drug approvals. Nature Reviews Drug Discovery 2021, 20, 85-90, 10.1038/d41573-021-00002-0.
  23. Asher Mullard; 2021 FDA approvals. Nature Reviews Drug Discovery 2022, 21, 83-88, 10.1038/d41573-022-00001-9.
  24. Daniela Bumbaca Yadav; Vikas K. Sharma; Charles Andrew Boswell; Isidro Hotzel; Devin Tesar; Yonglei Shang; Yong Ying; Saloumeh K. Fischer; Jane L. Grogan; Eugene Y. Chiang; et al.Konnie UrbanSheila UlufatuLeslie A. KhawliSaileta PrabhuSean JosephRobert F. Kelley Evaluating the Use of Antibody Variable Region (Fv) Charge as a Risk Assessment Tool for Predicting Typical Cynomolgus Monkey Pharmacokinetics. Journal of Biological Chemistry 2015, 290, 29732-29741, 10.1074/jbc.m115.692434.
  25. Henryk Mach; Sonia M Gregory; Alexis Mackiewicz; Sarita Mittal; Anita Lalloo; Marc Kirchmeier; Mohammed Shameem; Electrostatic interactions of monoclonal antibodies with subcutaneous tissue. Therapeutic Delivery 2011, 2, 727-736, 10.4155/tde.11.31.
  26. C. Jackisch; V. Müller; C. Maintz; S. Hell; B. Ataseven; Subcutaneous Administration of Monoclonal Antibodies in Oncology. Geburtshilfe und Frauenheilkunde 2014, 74, 343-349, 10.1055/s-0034-1368173.
  27. Ian K. Styles; Orlagh M. Feeney; Tri-Hung Nguyen; Daniel H.S. Brundel; David W. Kang; Renee Clift; Michelle P. McIntosh; Christopher J.H. Porter; Removal of interstitial hyaluronan with recombinant human hyaluronidase improves the systemic and lymphatic uptake of cetuximab in rats. Journal of Controlled Release 2019, 315, 85-96, 10.1016/j.jconrel.2019.10.040.
  28. Kenneth W. Locke; Daniel C. Maneval; Michael J. LaBarre; ENHANZE® drug delivery technology: a novel approach to subcutaneous administration using recombinant human hyaluronidase PH20. Drug Delivery 2019, 26, 98-106, 10.1080/10717544.2018.1551442.
  29. Marie A. Printz; Samuel S. Dychter; Emanuel P. DeNoia; Rena Harrigan; Barry J. Sugarman; Monica Zepeda; Jennifer Souratha; David W. Kang; Daniel C. Maneval; A Phase I Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Recombinant Human Hyaluronidase PH20 Administered Intravenously in Healthy Volunteers. Current Therapeutic Research 2020, 93, 100604, 10.1016/j.curtheres.2020.100604.
  30. O Shpilberg; C Jackisch; Subcutaneous administration of rituximab (MabThera) and trastuzumab (Herceptin) using hyaluronidase. British Journal of Cancer 2013, 109, 1556-1561, 10.1038/bjc.2013.371.
  31. Chris Wynne; Vernon Harvey; Christian Schwabe; Devonie Waaka; Christine McIntyre; Beate Bittner; Comparison of Subcutaneous and Intravenous Administration of Trastuzumab: A Phase I/Ib Trial in Healthy Male Volunteers and Patients With HER2-Positive Breast Cancer. The Journal of Clinical Pharmacology 2013, 53, 192-201, 10.1177/0091270012436560.
  32. Atsushi Ogata; Yasuhiro Kato; Shinji Higa; Keiji Maeda; Subcutaneous tocilizumab: recent advances for the treatment of rheumatoid arthritis. Expert Opinion on Drug Delivery 2019, 16, 639-648, 10.1080/17425247.2019.1618828.
  33. Paula Dakin; Stephen J. DiMartino; Haitao Gao; Jennifer Maloney; Alan J. Kivitz; Thomas J. Schnitzer; Neil Stahl; George D. Yancopoulos; Gregory P. Geba; The Efficacy, Tolerability, and Joint Safety of Fasinumab in Osteoarthritis Pain: A Phase IIb/III Double‐Blind, Placebo‐Controlled, Randomized Clinical Trial. Arthritis & Rheumatology 2019, 71, 1824-1834, 10.1002/art.41012.
  34. Hua, L.H.; Alvarez, E.; Foley, J.; Henry, R.G.; Brown, J.; Camacho, E.; Meng, X.; Ziehn, M.; Brown, B.; Greenberg, B.M.; et al. OLIKOS Study Design: Exploring Maintained Ofatumumab Efficacy in Relapsing MS Patients who Transition from Intravenous Anti-CD20 Therapy. Neurology 2021, 96 15 supplement, 4567.
  35. David M. Weinreich; Sumathi Sivapalasingam; Thomas Norton; Shazia Ali; Haitao Gao; Rafia Bhore; Bret J. Musser; Yuhwen Soo; Diana Rofail; Joseph Im; et al.Christina PerryCynthia PanRomana HosainAdnan MahmoodJohn D. DavisKenneth C. TurnerAndrea T. HooperJennifer D. HamiltonAlina BaumChristos A. KyratsousYunji KimAmanda CookWendy KampmanAnita KohliYessica SachdevaXimena GraberBari KowalThomas DiCioccioNeil StahlLeah LipsichNed BraunsteinGary HermanGeorge D. Yancopoulos REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19. New England Journal of Medicine 2021, 384, 238-251, 10.1056/nejmoa2035002.
  36. Hanne M. Kinnunen; Vikas Sharma; Luis Rodrigo Contreras-Rojas; Yafei Yu; Chlöe Alleman; Alavattam Sreedhara; Stefan Fischer; Leslie Khawli; Stefan T. Yohe; Daniela Bumbaca; et al.Thomas W. PatapoffAnn L. DaughertyRandall J. Mrsny A novel in vitro method to model the fate of subcutaneously administered biopharmaceuticals and associated formulation components. Journal of Controlled Release 2015, 214, 94-102, 10.1016/j.jconrel.2015.07.016.
  37. Hanne Kinnunen Bown; Catherine Bonn; Stefan Yohe; Daniela Bumbaca Yadav; Thomas W. Patapoff; Ann Daugherty; Randall J. Mrsny; In vitro model for predicting bioavailability of subcutaneously injected monoclonal antibodies. Journal of Controlled Release 2018, 273, 13-20, 10.1016/j.jconrel.2018.01.015.
More
Video Production Service