Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

TagedEnJournal of Pharmaceutical Sciences 112 (2023) 1492−1508

Contents lists available at ScienceDirect

TagedFiur TagedEn Journal of Pharmaceutical Sciences TagedFiur TagedEn

jo urn a l h om ep ag e : w ww.jp harms ci.o rg

Review

TagedH1Simulate SubQ: The Methods and the MediaTagedEn


TagedPDavid Li, Poh Yee Chow, Tzu Ping Lin, Celine Cheow, Zhuoxuan Li,
Matthias G. Wacker*TagedEn
TagedPNational University of Singapore, Faculty of Science, 4 Science Drive 2, 117544 Singapore
TagedEn
TAGEDPA R T I C L E I N F O TAGEDPA B S T R A C T

Article history: For decades, there has been a growing interest in injectable subcutaneous formulations to improve the
Received 16 August 2021 absorption of drugs into the systemic circulation and to prolong their release over a longer period. However,
Revised 25 October 2021 fluctuations in the blood plasma levels together with bioavailability issues often limit their clinical success.
Accepted 26 October 2021
This warrants a closer look at the performance of long-acting depots, for example, and their dependence on
Available online 30 October 2021TagedEn
the complex interplay between the dosage form and the physiological microenvironment. For this, biopredic-
TagedEnTagedPKeywords: tive performance testing is used for a thorough understanding of the biophysical processes affecting the
absorption of compounds from the injection site in vivo and their simulation in vitro. In the present work, we
Drug release
Dissolution testing
discuss in vitro methodologies including methods and media developed for the subcutaneous route of admin-
Performance testing istration on the background of the most relevant absorption mechanisms. Also, we highlight some important
Biorelevant knowledge gaps and shortcomings of the existing methodologies to provide the reader with a better under-
Biopredictive standing of the scientific evidence underlying these models.
Depot formulation © 2021 American Pharmacists Association. Published by Elsevier Inc. All rights reserved.TagedEn
Long-acting injectables
Parenterals
Drug delivery
Drug absorption
ADME
In vitro-in vivo correlation (IVIVC)
Lymphatic uptake
Subcutaneous (SC)
Absorption
Non-oral complex drugs TagedEn

TagedH1IntroductionTagedEn agedEnTPSC delivery can be a convenient and effective alternative.1−3 Also,


the SC tissue often serves as a depot, leading to prolonged-release
TagedPIn recent years, drug formulations for subcutaneous (SC) injec- intervals and reduced dosing frequency while avoiding extensive
tion have experienced a surge in demand due to a number of first-pass metabolism.1 Furthermore, when combined with suit-
advantages as compared to other dosage forms. From a drug able devices, the formulation can be administered by the patient
development perspective, drugs with poor aqueous solubility, and does not require healthcare professionals. They increase
permeability, or stability issues in the gastrointestinal tract are patient compliance and reduce hospitalization costs.1,4 Although
often limited by their peroral bioavailability. For these candidates, the SC route of administration offers exciting opportunities in
drug delivery, pharmaceutical development is accompanied by
many challenges. So far, a widely recognized and validated pre-
TagedEnAbbreviations: DR, Dispersion Releaser; ECM, Extracellular matrix; FBS, Fetal bovine clinical model to predict the absorption and bioavailability in
serum; HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IVIS, In vivo imag- humans has yet to be established. Biopredictive in vitro perfor-
ing system; IVIVC, In vitro-in vivo correlation; MbCD, Methyl-b-cyclodextrin; NMP, N-
mance assays often fail to mimic the complexity of the in vivo
methyl-2-pyrrolidone; PBS, Phosphate buffered saline; PCL, Poly(e-caprolactone); PEG,
Polyethylene glycol; PLA, Poly(D,L-lactic acid); PLA-PCL, Poly(D,L-lactide-co-e-capro- absorption process and safety characteristics.5,6 Also, many of the
lactone); PLGA, Poly(D,L-lactic-co-glycolic acid); PLLA, Poly(L-lactic acid); PVA, Polyvi- known influences on drug absorption come with high uncertainty
nyl alcohol; RES, Reticuloendothelial system; SBF, Simulated body fluid; SC, due to the significant interspecies and interindividual differences
subcutaneous(ly); SCISSOR, Subcutaneous Injection Site Simulator; SDS, Sodium as only a small minority of literature studies are currently based
dodecyl sulphate; SSIF, Simulated subcutaneous interstitial fluid.
TagedEn* Corresponding author.
on larger animals or humans. Most information on the SC route
E-mail address: matthias.g.wacker@nus.edu.sg (M.G. Wacker). of administration is obtained from rodents (Fig. 1, left).TagedEn

https://doi.org/10.1016/j.xphs.2021.10.031
0022-3549/© 2021 American Pharmacists Association. Published by Elsevier Inc. All rights reserved.
TagedEn TagedFiur TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1493

Figure 1. Of 295 articles listed on Pubmed (2011-2021) under the keywords “subcutaneous injection” and “formulation”, we evaluated a total of 136 randomly selected full-
research articles for the species used to investigate drug delivery of poorly soluble compounds (left), and the studied influences affecting this process in the SC tissue (right). The
number of review articles in this time interval was 36.TagedEn

TagedPThese knowledge gaps can put drug development but also the test TagedEnPthe SC tissue. Therefore, metabolic stability has to be considered as
subjects at risk.7 They complicate translation to the clinical stages.5 one of the parameters of interest as well.TagedEn
Therefore, a better understanding of the various influences of the TagedPPutting more focus on the administration site itself, the SC tissue
dosage form on bioavailability and safety characteristics is required. is characterized by the presence of mostly adipocytes, with access to
Improved prediction of events such as the initial burst release or the the blood capillaries and lymphatic vasculature,15 while the intersti-
lymphatic uptake of fine particles into the systemic circulation would tium, amongst other components, contains plasma proteins at a
improve the translation from bench to bedside and ensure that more lower concentration as compared to the vascular system.16 Being
candidates meet the regulatory and therapeutic requirements.8−11TagedEn more poorly perfused than the intramuscular environment, the SC
TagedPThe interspecies differences in the SC environment between tissue enables drugs to be formulated into SC depots and sustained-
humans and animals result in conflicting outcomes and unpredict- release formulations that are slowly absorbed into the systemic circu-
able pharmacokinetics.12,13 This diversity is reflected by the perfor- lation by passive diffusion.17 An illustration of the administration site
mance assays as well. In the present work, we critically review the and the most important mechanisms involved in the in
current knowledge on absorption processes relevant for the SC route vivo absorption process is presented in Fig. 2, providing the base for a
of administrations. Emphasizing the physiological processes involved more thorough discussion in the following sections.TagedEn
in drug release and absorption, we highlight challenges and consider-
ations in the development of biopredictive performance assays. Fur- TagedH2Lymphatic UptakeTagedEn
ther, we lay open critical knowledge gaps and discuss open questions
in this research area.TagedEn TagedPWhile many biopharmaceuticals such as large proteins or nucleic
acids are characterized by a high molecular weight, poorly soluble
TagedH1Influences on Drug AbsorptionTagedEn compounds are often formulated into particles, vesicles, or micelles
to improve their solubility at the injection site. This includes the
TagedPTo successfully develop drug products for SC injection, a careful encapsulation into liposomes,18 microsizing or nanosizing of the
evaluation of all physicochemical characteristics involved in the crystalline drug,19 or the covalent modification of drugs by attaching
absorption process is required. Critical material attributes can have a them to drug carriers such as dendrimers. The latter sometimes
strong impact on the interactions of the dosage form with the physio- includes the use of macromolecules.18 Generally speaking, macromo-
logical microenvironment. They include parameters such as the lecules with a molecular weight larger than 1 kDa have limited
aqueous solubility in physiological fluids, the buffer capacity at the mobility in the SC tissue. The absorption into blood vessels is much
injection site as well as physical or chemical stability. Also, the parti- slower as compared to smaller molecules. While for molecules with a
cle or molecular size of the formulation can have a strong influence size below 100 nm, absorption into the lymphatic system is a prefera-
on the mobility of the drug in the interstitial space.14 To provide a ble pathway, from where they eventually drain into the systemic cir-
basis for our critical analysis, we evaluated articles from current liter- culation at the thoracic lymph duct.16 In this section, we will look at
ature dealing with SC delivery of poorly soluble compounds (Fig. 1, the factors that affect the lymphatic uptake and lymph node localiza-
right). Among all the influences on drug absorption, the rate or extent tion, which may consequently influence the amount of drug that can
of absorption, release kinetics of drugs from their delivery system, reach the systemic circulation. Firstly, a process that affects whether
the interaction of formulations with the lymphatic system as well as the drug reaches the systemic circulation from the lymphatic system
the biocompatibility of the material were explored. For most biophar- is the susceptibility of these particles to phagocytosis by the macro-
maceuticals, the chemical stability of the compound together with phages from the RES. Foreign particles in the body are detected by
the uptake by the reticuloendothelial system (RES) rather than the macrophages through their particle size, surface charge, and hydro-
aqueous solubility are limiting bioavailability. This includes larger phobicity when they differ largely from the endogenous compounds,
proteins such as antibodies but also nucleic acids and peptides. While as part of the immune system to defend against pathogens. Thus, if
large proteins and nucleic acids have a higher risk of being recog- these signature recognition points can be concealed, phagocytosis
nized by the immune system, peptides are sensitive to enzymes in can be reduced. Using PEGylation, a process involving the attachment
TagedEn TagedFiur
TagedEn1494 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

Figure 2. Illustration of the subcutaneous injection site including mechanisms involved in the absorption process. This illustration was created with BioRender (www.biorender.com).
TagedEn

TagedEnPof polyethylene glycol (PEG) molecules to the particle surface, lym- TagedEnPapproximately 100 nm,16,32−34 with another study specifying that
phatic drainage can be improved as it can reduce the susceptibility of the optimal size for lymphatic uptake is between 10 to 50 nm.35 Pri-
nanoparticles, for example, to phagocytosis by macrophages. The marily rodent models were used to study lymphatic uptake, except
hydrophilic surface structure prevents opsonization by reducing the for one review34 which referenced a paper using a porcine model.36
binding of proteins that stimulate immune responses. It has been The porcine skin is structurally more similar to the human skin as
demonstrated that the PEGylation of various drug carriers such as lip- compared to that of rodents.37 Since the studies using rodents largely
osomes, solid lipid nanoparticles, and dendrimers was able to reduce concur with the results of the study using a porcine model, the con-
phagocytic uptake.20−22 Besides PEGylation, a similar outcome was clusion made with the porcine model can serve as supporting evi-
also achieved through the incorporation of GM1 gangliosides into the dence that the optimal size for lymphatic uptake may be between 10
composition of liposomes,23 due to the presence of sialic acid which and 100 nm. Though more research is still required to directly corre-
was also able to impart hydrophilicity onto the nanoparticle surfaces late this data to humans.TagedEn
for RES evasion.24TagedEn TagedPWhile we can assume that an optimal size can lead to an increase in
TagedPOther parameters that affect the surface structure and reduce lymphatic uptake, aforementioned modifications and exceptions need
hydrophobic interactions are surfactants such as poloxamers and to be remembered and its general impact on the overall absorption into
poloxamines. They aid drug molecules in their movement through the blood circulations needs to be discussed. On one hand, a study in
the hydrophilic water channels in the interstitium.21,25,26 With mice highlighted that the size and surface modification of liposomes
increased diffusion through the interstitium, more formulations are with PEG altered drug absorption. Smaller liposomes and the presence
thus likely to enter the lymphatic system, and subsequently, drain of PEG on the surface led to higher blood levels as compared to larger
into the systemic circulation.TagedEn liposomes and liposomes without PEG, respectively.31 On the other
TagedPThe particle or molecular size of drugs and drug carriers affect hand, another study using rats concluded that lymphatic uptake is
lymphatic uptake as well. Larger liposomes commonly lead to a low- unlikely to have a significant impact on systemic absorption,21 concur-
ered lymphatic uptake. This is likely due to the limited size of water ring with Oussoren et al. that higher lymphatic uptake does not neces-
channels in the interstitium. They create a filter that particles have to sarily correlate with an increase in blood concentration.38TagedEn
pass through to reach the lymphatic system.27 While molecules TagedPWith regards to liposomes, the drainage of drugs and their formu-
smaller than 10 nm are more likely to be absorbed through blood lations into the systemic circulation may furthermore be affected by
capillaries,28 the upper cut-off size for lymphatic uptake has been the exact injection site, liposomal bilayer fluidity, and the type of lip-
reported differently in a few studies, with two review papers ids used in the formulation.27,39 Many of the abovementioned studies
referencing studies using rats and mice in their analysis concluding used rodents to determine the impact of the lymphatic system on
the cut-off size to be 100 nm and 150 nm each,29,30 while primary lit- drug absorption, some of them with conflicting outcomes. One reason
erature using mice reported a cut-off size between 110 nm to for this may be the aforementioned injection site. Allen et al. injected
120nm.31 Overall, there seems to be a consensus that the cut-off is at liposomes into the neck or upper back just below the neck of the
TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1495

TagedEnPanimal.31 In two other studies,21,31 the drug formulations were TagedEnPbioavailability. On the other hand, the presence of adipose tissue con-
injected into the hind legs and dorsal side of the foot, respectively. tributes to a stronger partitioning effect.46,59 In summary, tissue
Overall, the dorsolateral areas of the neck or back are among the pre- retention is a strong impact on absorption that needs to be consid-
ferred sites for SC injection in rats.40 As such, results from the earlier ered.TagedEn
study resulting in higher blood levels may be more reliable for our
interpretation.31TagedEn TagedH2Release KineticsTagedEn
TagedPIn conclusion, a carefully selected particle size, as well as surface
modifications with PEG, seem to increase absorption from the SC tis- TagedPFor many injectable formulations, the drug release from the car-
sue into the blood circulation, potentially through lymphatic uptake. rier limits the bioavailability of the compound in systematic circula-
This is reflected by the most successful formulation strategies as well. tion. The mechanism of release can be governed by various processes
Surface PEGylation has become a common strategy to improve the such as the degradation or erosion of the carrier, the swelling, and
stability and absorption of large and medium-size proteins such as wetting of the matrix material, or the diffusion of excipients and the
interferon.41,42 Lymphatic drug exposure itself remains complex and drug through the SC tissue.8,60,61TagedEn
whether the abovementioned animal models are able to predict lym- TagedPDrug diffusion is affected by the concentration gradient between
phatic distribution in humans is a topic that still requires more the injection site and the physiological microenvironment. Conse-
research. Though, with the rise of lymphatic imaging techniques, pre- quently, the partitioning between adipocytes and the interstitial
viously invasive methods can be replaced to gain more insight and space as well as the plasma protein binding can play an important
enable more representative studies designed on humans in the near role. Critical changes in the degradation or diffusion processes will
future. In recent years, such techniques have already been used to affect the release rate. With regards to polymeric carriers and lipo-
study the biodistribution of drugs from SC depots in animals43 as somes, the degradation may occur through hydrolysis or enzymatic
well as in humans.44TagedEn degradation into smaller units of monomers or oligomers.60 The deg-
radation rate depends on the composition and structure of the matrix
TagedH2Tissue Retention and Molecular MobilityTagedEn in various types of drug carriers.33,62,63 For example, the use of chito-
san/poly(L-lactic acid) (PLLA) composite microspheres increased the
TagedPIn addition to the lymphatic drainage of particles and mole- rate of degradation of the microspheres by hydrolysis. The positively
cules, the bioavailability of therapeutics is also affected by the charged chitosan led to pore formation in the microparticle system.
retention of the drug in the SC tissue. Particles in the micrometer This increased the surface area and thus the access of water mole-
range are known to remain at the injection site and exhibit poor cules to the polymeric matrix.62 In other cases, the degradation rate
mobility while smaller particles undergo a distribution process was affected by the type of drug incorporated in the drug carriers as
towards the systemic circulation. Diffusion properties are strongly well.64 The pore size of microparticles composed of poly(lactic-co-
affected by the chemical structure and the excipients used in the glycolic acid) (PLGA) is commonly controlled by the ratio of lactide to
formulation. Introducing a negative charge to the carrier or the glycolide with a strong influence on water diffusion, hydrolysis, and
molecule strengthens the repulsion forces between the formulation release behavior.65,66 When hydrophilic compounds were incorpo-
and the negatively charged glycosaminoglycans or chondroitin rated into these drug carriers, they increased the rate of hydrolysis
present in the SC tissue.45,46 Chondroitin has two ionizable groups by facilitating the diffusion of water into the matrix. This has been
per repeated unit, comprising both carboxylate and sulfate moie- observed for small-molecular drugs aspirin and ibuprofen.64 Con-
ties (carboxylate pKa 3-5, sulfate pKa 1.5-2).47 Hyaluronic acid versely, an increase in the ratio of hydrophobic lactide to glycolide in
exhibits a pKa of 2.9 at physiological pH. Although hyaluronan rep- a PLGA matrix reduces water diffusion, and thus hydrolytic degrada-
resents only 1% of the concentration of collagen in the skin, the tion of the copolymer.64 Degradation of poly(D,L-lactic acid) (PLA)
fluid exclusion volume potential is much higher as compared to and PLGA involves autocatalysis in a low pH environment within the
collagen.48,49 For negatively charged antibodies reduced endocyto- polymeric matrix following hydrolysis, due to the release of acidic
sis and higher bioavailability have been reported.50 On the con- monomers upon hydrolysis of the polymers.67TagedEn
trary, positively charged molecules often bind to the extracellular TagedPThe fluid volume and buffer capacity in the SC tissue are very lim-
matrix, leading to a delay in the absorption rate.51−53TagedEn ited. Therefore, the impact of the formulation on the pH is more pro-
TagedPHydrophilic drugs tend to appear in the blood plasma more rapidly nounced as compared to other administration routes. Other factors
due to their improved solubility and rapid diffusion through the SC that can contribute to a low pH environment would be the use of
environment. For many biopharmaceuticals such as small peptides and acidic drugs or polymers in the formulation.8 A reduced pH has been
medium-size proteins, this increases absorption and bioavailability.54,55TagedEn associated with a change in the release kinetics and is partially
TagedPFor larger proteins such as antibodies, the molecular weight leads affected by tissue changes. The effect was reported for implant for-
to significant retention at the administration site. The three-dimen- mulations composed of PLGA.68TagedEn
sional network formed by the extracellular matrix (ECM) reduces the TagedPSo far, there is limited evidence for the autocatalysis of basic
mobility of the molecule and, hence, allows enzymes to degrade the drugs.65,69,70 However, porosity8 and dimensions of drug carrier71
drug before it appears in the blood plasma. The abovementioned have an influence on the diffusion of acidic degradation products out
modification of proteins with PEG is a common strategy to improve of their matrices. The reduced diffusion causes an accumulation of
the stability of compounds. However, previous investigations acidic compounds and enhances biodegradation. These effects have
reported minor alterations in the molecular weight (PEG30 to PEG40) been reported for a variety of PLGA and PLA polymers.72 As such,
having a strong impact on SC absorption.56 This change is due to the autocatalysis contributes more significantly to the degradation of
altered tissue retention and diffusion rate and affects particles as larger and non-porous microparticles73 and implants72. Furthermore,
well.57 One major difference between the SC and the intramuscular an evaluation of different mathematical models highlighted that
route of administration is the degree of vascularization. As a conse- accounting for autocatalysis increased the predictability of drug
quence, the diffusion pathway of SC formulations is considerably lon- release kinetics.71TagedEn
ger resulting on average in lower bioavailability. A similar effect is TagedPThe use of chitosan in chitosan/PLLA composite microspheres
found for different injection sites.58TagedEn increased the buffer capacity, compensating for the hydrolysis of
TagedPHydrophobic interactions often lower the absorption.46 On one PLLA to lactic acid. This led to a slower decrease in pH. Despite this
hand, the poor aqueous solubility of these compounds decreases reduction in autocatalysis, the rate of degradation of composite
TagedEn1496 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

TagedEnPmicrospheres was still higher as compared to pure PLLA micro- TagedPInterestingly, the release of histamine and serotonin following the
spheres. The availability of water due to the improved pore formation injection trauma was found responsible for reduced absorption. This
outweighs the influence of pH in vitro. An in vivo experiment con- phenomenon was observed in rats only.87 However, histamine indu-
ducted in rodents further confirmed the preferential dissolution of ces vasodilation of small arteries88 and increases permeability89 in
chitosan over PLLA in a physiological environment.62TagedEn the SC tissue of humans. Still, the effects of histamine on the vascular
TagedPThe next process that affects drug release is drug diffusion. It tone vary between different species and can hardly be predicted
can be delayed through stronger drug-drug interaction, drug- based on rodent studies alone.88 In some studies, antihistamines
matrix interactions, and larger particle sizes.65 Varying the have been used to reduce allergy-related side effects following the
strength of the interactions between the drug and its carrier has injection of insulin.90 So far, there is limited information on the
been demonstrated to vary the rate of release.63,74,75 However, the effects of these treatments on drug absorption. However, future
parameter depends on multiple interactions of the drug delivery research could consider monitoring the effect of antihistamines more
system, and measuring the impact of the inherent binding forces is carefully. They are often used as a co-treatment and could provide
challenging. A study comparing the drug release of beta-blockers unexpected insights.TagedEn
with similar molecular weights and pKa, but varying in their lipo- TagedPOn another note, long-term exposure to foreign particles, in
philicities showed that despite the difference in drug release rate, general, can lead to long-lasting inflammation causing the forma-
the difference in the rate of absorption was not statistically tion of fibrotic tissue or a fibrous capsule. This is especially pro-
significant.76TagedEn nounced for non-degradable materials.91 It is generally seen as an
TagedPOne major shortcoming of SC drug formulations is the occur- indication of poor biocompatibility of the material that occurs as
rence of the “burst effect”. Sometimes, it is affected by the biodeg- part of an inflammatory response.92 This has been observed for
radation of the drug carrier and the diffusion behavior of the implanted microspheres,78 devices,93 but also proteins or nucleic
drug. A very common explanation is the heterogeneous distribu- acids.94,95 The effect occurs two to three weeks after the adminis-
tion of drug molecules in the matrix material, where each phase tration of implants96 and was found to pose a barrier to drug
comes with a distinct release and absorption behavior.77 Drug at absorption.96 Therefore, most implants and formulations change
the surface of the carrier is released much more rapidly than their absorption behavior one to four weeks after implantation.97
drug encapsulated into the core.63,78 The distribution widely The thickness of the capsule increases with the implant thickness
depends on the encapsulation process but may also be affected or diameter. This translates into a longer diffusion distance and
by the storage conditions.79 This effect can be countered through greater diffusion resistance respectively. Although the formation of
the creation of additional diffusion barriers.10 However, the burst such a capsule may lead to prolonged-release intervals, local fibro-
effect that is often held responsible for an initial increase in the sis must be considered a serious and widely unpredictable adverse
blood plasma concentration is only one puzzle piece of the initial reaction. The expected interpatient variability and physiological
absorption and can overlap with lymphatic uptake or other consequences of the fibrosis (e.g., formation of scars) do not allow
parameters discussed in this article.TagedEn the use of this effect in drug delivery.TagedEn
TagedPAt last, PEGylation of biomolecules may also cause activation of
TagedH2Biocompatibility and Responses of the Immune SystemTagedEn immune cells. The responses to such an adverse reaction are often
unpredictable as well and require additional immunogenicity assays
TagedPThe biocompatibility of formulations sometimes has a certain influ- to elucidate the pharmacological responses to the delivery system.
ence on drug absorption. Poor biocompatibility potentially induces These assays are not within the scope of this review article. Interested
inflammation and may lead to drastic changes in the SC microenviron- readers are referred to summaries provided by Dingmann and Balu-
ment. These reactions are often caused by formulations with high con- Iyer98 or Jarvi and Balu-Iyer.99TagedEn
centrations of solubility enhancers. Co-solvents or complexation agents
are amongst the most critical excipients as common aims of formula-
tion development include the solubilization of poorly soluble com- TagedH2Metabolic StabilityTagedEn
pounds.80 For those candidates, precipitation may occur when
supersaturated solutions are injected into the physiological environ- TagedPWhile most chemical entities are characterized by high molecular
ment. The diffusion of excipients such as water, buffer salts, solvents, stability, biodegradable drugs such as peptides, proteins, or nucleic
and co-solvents, from the injection site into the surrounding tissues acids undergo a rapid metabolism in the SC tissue. In this context, the
reduces the physical stability of the formulation14 and sometimes time required by the drug molecule to reach the nearest blood vessel
causes rapid crystallization of the drug. As a consequence, inflammation and drug degradation rate are interdependent parameters with a
at the injection site may occur. However, supersaturated solutions do strong impact on bioavailability. Metabolism can be widely driven by
not necessarily precipitate. The induction time, which is the time before the proteases secreted into the SC tissue.100 They include serine pro-
precipitate formation, is negatively correlated to the extent of supersat- teases,101 cathepsin-B, and collagenase-like peptidase proteases,102
uration.80 Phospholipid formulations such as liposomes81 but also high as well as aminopeptidases.103 The stability of glucagon-like peptide
concentrations of colloid stabilizers with high affinity to the particle 1 analogs in the SC has been investigated in more detail.104 A direct
surface are likely to reduce such precipitation effects.TagedEn correlation between degradation of the drug and bioavailability was
TagedPInflammation can also be caused by penetration trauma, pH found.104 Other drugs such as tacrolimus33 undergo non-enzymatic
changes in the SC tissue, charge interactions, the high osmotic activ- chemical reactions including hydrolysis or oxidation.105,106 As men-
ity of the dosage form, or the released drug itself.82−85 The latter can tioned in the previous sections, the metabolic stability of proteins
happen with biopharmaceuticals such as larger proteins or nucleic and peptides can be improved by conjugating the drug molecule
acids in particular. Though in another example, the release of naltrex- with PEG41,42 or hydroxyethyl starch.107,108 So far, PEGylation tech-
one from PLGA-based drug carriers in rats and rabbits has led to a sta- nology was most successfully used in the design of novel drug candi-
tistically different inflammatory response as compared to placebo. dates.109 Alternatively, the compound can be encapsulated into
Naltrexone was the main contributor to the inflammatory response delivery systems such as micro-/nanoparticles110 or liposomes.111
rather than the degradation products of PLGA.83 The extent of inflam- Predicting the impact of novel formulation strategies on the release,
mation was reduced when the drug was administered at a lower stability, and bioavailability of drug molecules is one of the most
concentration.86TagedEn important goals of biopredictive performance testing.TagedEn
TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1497

TagedH1Biopredictive Methods and MediaTagedEn TagedEnPinjectables, more complex and biorelevant media reflecting the
microenvironment at the injection site may be required. Simple
TagedPAt present, a wide variety of in vitro assays are being developed to buffer systems sometimes fail to mimic the most important processes
predict the in vivo performance of SC drug formulations.33,43,58,78,112 involved in the release and may not be suitable to predict the in vivo
−115
Commonly, biorelevant methods provide an accurate reflection performance.TagedEn
of the physiological environment including the medium composition,
fluid volume, and hydrodynamics. They often contain proteins or TagedPIon Species, Concentration, and Protein ContentTagedEn
ions present at the injection site. For comparison, biopredictive meth- TagedPFor some formulations, specific components of the medium such
ods demonstrate a partial ability to predict human in vivo perfor- as enzymes135 or certain ion species136,137 can have a strong impact
mance and may contain non-physiological ingredients such as on the release of the drug product. Consequently, this influence
surfactants or stabilizers. More than an accurate reflection of the in should be reflected by the in vitro assay. Simulated body fluid (SBF),
vivo situation, they trigger selected mechanisms of release such as for example, comprises Ca2+ and Mg2+ cations in physiological con-
the dissolution of the drug or the degradation of the matrix material. centrations. Hence, this fluid has been used for release and degrad-
Most release assays for SC formulations do not achieve a high level of ability studies of mesoporous silica microparticles where the cation
biorelevance. Sometimes depot formulations with an in vivo release concentration plays a significant role.136,137 Although it does not spe-
over several days, weeks, or months are rather tested under acceler- cifically mimic the SC environment, SBF was used in a variety of bio-
ated conditions.33,113,116,117 Quality control methods can use this to activity, degradation, and dissolution studies focusing on these
discriminate quality differences between different batches of the applications.138−140 Kokubo et al. postulated that ion concentrations
same dosage form. They reflect the rate-limiting release mechanism and chemical composition reflect the physiological situation in
but have a more limited predictive power for in vivo performances.118 human plasma. This gives it a certain advantage as compared to other
Shen and Burgess present further information on accelerated in vitro fluids.141TagedEn
release testing methods for extended-release parenteral dosage TagedPPhosphate buffered saline (PBS) is another common medium. As
forms in general in their review article.119TagedEn compared to human plasma and SBF, it comprises a much higher
TagedPValidation of the in vitro assay can be accomplished by means of in phosphate concentration and generally does not contain Ca2+ or
vitro-in vivo correlation (IVIVC). These mathematical relationships Mg2+.142 Nonetheless, PBS is widely applied in the simulation of
between the in vitro and the in vivo data obtained from the same for- extracellular fluids. Compared to the other phosphate buffers, it
mulation commonly use bioavailability as a surrogate parameter for has the advantage that the composition is well-established. This
the expected in vivo response. The absorbed fraction of the drug can allows a direct comparison between different setups. Another
be calculated by deconvolution from the plasma concentration-time advantage of PBS is that the medium matches the osmolarity of
profile using model-dependent techniques such as the Wagner-Nel- human body fluid. It contains sodium and chloride ions in physio-
son or the Loo-Riegelmann method. They define a theoretical frame- logical concentrations.133 Another medium is Hanks’ balanced salts
work (the mathematical model) to isolate the absorption process. solution (HBSS). It comprises ion species and concentrations more
The evolution of computational methods in pharmaceutical develop- comparable to SBF, while also containing glucose. For SBF and
ment has resulted in the integration of physiological parameters to HBSS, multiple versions have been published, depending on the
create more realistic simulations of the in vivo setting. Physiologi- technical requirements of the assay. For example, Iyer et al. postu-
cally-based biopharmaceutics modeling (PBBM) is a discipline lated a modified HBSS for in vitro release testing of naltrexone
involved with the explanation of the absorption phase.TagedEn implants.133 Phenol red, a usual component of HBSS was removed
TagedPHowever, a significant number of influences on drug absorption due to an interference with the analytical method. Also, he used 4-
are not solely controlled by the formulation design. Therefore, new (2-hydroxy ethyl) piperazine-1ethanesulfonic acid (HEPES) as a
standards must be defined for the application of computer models in buffer system, replacing the carbonate to reach a much higher
the development of IVIVCs.118TagedEn buffer capacity. This was to conduct the measurement under accel-
TagedPIn the following, we will discuss the release media and methods erated conditions. Primocin was used as a preservative. In sum-
applied in the simulation of the SC tissue on the background of mary, SBF, PBS, and HBSS exhibit some characteristics commonly
parameters affecting drug absorption.TagedEn found at the injection site but, with regards to their biorelevance,
can be considered a simplified version of the physiology. SBF and
TagedH2Release MediaTagedEn HBSS are closer to the composition of physiological fluids. This can
play an important role whenever the in vivo release is triggered by
TagedPMedium development often involves a compromise between the a specific ion concentration or species.TagedEn
existing knowledge on the microenvironment in vivo and the techni- TagedPSerum proteins affect the release rate but, more often, have an
cal requirements of release testing. The most common parameters impact on chemical quantification methods and reproducibility of
are pH, buffer capacity, and ion concentration at the injection site. the in vitro assay as well. Gao et al. explored the addition of pro-
They affect the ionization of compounds or excipients and can have a teins, namely fetal bovine serum (FBS), to a release medium
strong impact on the release. Water, as a release medium, has no intended for the simulation of the SC route of administration. This
buffer capacity and is thus, the most unreliable medium for the test- simulated subcutaneous interstitial fluid (SSIF) leans on the com-
ing of compounds and formulations affected by pH. However, even ponents and concentrations found in interstitial fluids instead of
for those formulations insensitive to such changes, there is a general human blood plasma, thus representing a biorelevant medium
consensus to evaluate the drug release under conditions reflecting at more specific for the SC injection site.58 This is even more relevant
least some characteristics of the administration site.112TagedEn because, due to the limited permeability of the capillary mem-
TagedPA selection of the most common release media applied in release brane, the protein concentration in the interstitial fluid is lower as
testing of SC drug formulations is presented in Table 1. Most in vitro compared to the blood.143 Nonetheless, serum proteins can have a
studies were carried out using a buffer system at pH 7.4, reflecting stabilizing effect on particle dispersions, counteract agglomeration
the physiological pH of the SC tissue. Commonly, phosphate buffers and change the release rate. Table 2 summarizes the ion concentra-
with concentrations ranging from 10 mM120 to 100 mM75,121,122 are tions and protein contents of blood plasma, interstitial fluid, and
used. With a rising number of poorly soluble compounds entering the abovementioned media. Recently, the compositions of various
the global healthcare market18 and their formulation into long-acting media simulating the SC route have been reviewed. The authors
TagedEn1498 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

TagedEnTable 1
Selected release media often used in vitro for the simulation of the SC environment.

Release media Notable modification Drug Formulation Refs.


75
Phosphate buffer (pH 7.4) 100 mM, Dexamethasone Core-crosslinked polymeric micelles
15 mM sodium chloride
75
100 mM, Paclitaxel Core-crosslinked polymeric micelles
15 mM sodium chloride,
Tween 80
121
100 mM, Nalmefene PLGA microspheres
sodium azide,
SDS
120
10 mM, Cytochrome c PLGA microspheres and implants
PVA,
sodium azide
122
100 mM, 4-benzoyloxy-2,2,6,6- In situ forming PLGA/NMP implants
tetramethylpiperidine-1-oxyl
123
50 mM, Buserelin PLGA implant
benzalkonium chloride,
sodium azide
117
67 mM Insulin Lipid implant
124
PBS (pH 7.4) Naproxen Nanosuspension
125
Bupivacaine Lipid-polymer hybrid nanoparticles
126
Tween 80 Oleanolic acid Liposomes
127
Carbonic anhydrase PLGA microspheres
127
Bovine serum albumin PLGA microspheres
128
Tween 20, Naltrexone PLGA microspheres
sodium ascorbate
10
Ibuprofen Composite PLGA microparticles
incorporating PCL nanoparticles
129
Dexamethasone PLGA microspheres mixed with
crosslinked hydrogel
130
Sodium azide Thymosin alpha 1 In situ forming PLGA implants
131
Tumor necrosis factor In situ forming PLGA implants
6
HEPES buffer (pH 7.4) 10 mM Dexamethasone Liposomes
113,132,133
Modified Hanks’ balanced HEPES buffer, Naltrexone PLA-PCL implant
salts solution (pH 7.4) primocin etc.
134
SBF (pH 7.4) SDS Toremifene citrate, Silica gel microspheres
dexmedetomidine HCl
58
SSIF FBS, Medroxyprogesterone acetate Microparticles
Pen-StrepÒ
33
FBS, Tacrolimus PLGA and PLA microspheres
MbCD
PBS = Phosphate buffered saline; SBF = Simulated body fluid; SSIF = Simulated subcutaneous interstitial fluid; HEPES = 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid;
SDS = Sodium dodecyl sulfate; PVA = Polyvinyl alcohol; FBS = Fetal bovine serum; MbCD = Methyl-b-cyclodextrin; PLGA = Poly(D,L-lactic-acid); PCL = Poly(e-caprolactone);
NMP = N-methyl-2-pyrrolidone; PLA = Poly(D,L-lactic acid); PLA-PCL = Poly(D,L-lactide-co-e-caprolactone).

TagedEnPoutlined the importance of an actual measurement of the physio- TagedEnPperformances may be a more reliable benchmark than the varying
logical composition of the SC fluid.144 Although a review of the compositions measured in animals or humans.TagedEn
data available has been timely, their analysis follows the common
narrative that today's biorelevant media offer an exact reflection of TagedPSurfactants, Solubilizers, and PreservativesTagedEn
the physiological environment. However, even for the well-estab- TagedPFor many injectables, accelerated conditions are a key issue in the
lished media simulating the peroral route, a balance between bio- development of new performance assays. An evaluation of the release
prediction and biorelevance was necessary.145 In this context, with over several days, weeks, or months is affected by the chemical and
regard to the SC route of administration, the poor standardization microbiological stability of the simulated microenvironment. This
of the administration site (e.g., upper arm, abdomen, thigh) and weakness not only affects biorelevant media but all biologically
difficulties in sample extraction must be taken into consider- inspired in vitro systems and explains the relative insignificance of
ation.144 Therefore, the capability of the medium to predict in vivo three-dimensional cell models in the evaluation of depot

TagedEnTable 2
Relevant ion concentrations and protein content of blood plasma, interstitial fluid, and media used for in vitro release and degradation studies of SC formulations.

Cations [mM] Anions [mM] Proteins [g/L]


+ + 2+ 2+ 2 2
Medium Na K Ca Mg Cl HCO3 HPO4 SO4

Blood Plasma146 142 5 2.5 1.5 103 27 1 0.5 60-80147


Interstitial Fluid58 143 4 1.3 0.7 115 28 1 0.5 26
PBS146 157 4.5 - - 140 - 10 - -
SBF146 142 5 2.5 1.5 148.8 4.2 1 0.5 -
HBSS148 142.8 5.8 1.3 0.9 146.8 4.2 0.3 0.4 -
SSIF58 136 3.9 1.3 0.5 114.9 20.6 1 0.5 25 (FBS)
FBS = Fetal bovine serum; HBSS = Hanks’ balanced salts solution; PBS = Phosphate buffered saline; SBF= Simulated body fluid; SSIF = Simulated subcutaneous interstitial fluid.
TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1499

TagedEnPformulations. Common additives include antioxidants such as sodium TagedPIn the past, hydrogel assays have been developed, to simulate the
ascorbate128 to prevent drug degradation, anti-adherents such as resistance of the physiological environment.117,153 A certain intersti-
polyvinyl alcohol120, and, more frequently, preservatives such as tial pressure resulting from constant muscle movement together
sodium azide,120,121,123,130 benzalkonium chloride,123 penicillin- with the interconnected network of the ECM may affect the release
streptomycin (Pen-StrepÒ ),33,58 and primocin113,132,133 when release of SC injectables. To simulate this interstitial pressure, among others,
testing is done over an extended period.TagedEn elastic agarose gels43 but also ECM components such as hyaluronic
TagedPFurthermore, to assure a complete release process, many media acid and collagen have been used.117,153TagedEn
are supplemented with solubilizing agents.149 They comprise surfac-
tants or cosolvents to accelerate the dissolution process and enable TagedPTemperatureTagedEn
sink conditions.33,75,121,126,128,134 Surfactants do not reflect the physi- TagedPAnother important parameter in the development of the release
ological conditions at the administration site. Still, they can allow a assay is the temperature. Currently, there is no common standard for
more accurate prediction of the in vivo performance when absorption the in vitro testing of SC drug formulations. Even the most recent USP
is mainly limited by the dissolution.TagedEn chapter <1001> In Vitro Release Test Methods for Parenteral Drug Prep-
arations provides no guidance in the area of SC drug formulations.
TagedPMedium Composition and CharacteristicsTagedEn While 37 °C is often regarded as the core temperature of the human
TagedPOther aspects to be considered are the medium composition, vol- body, temperatures in the SC tissue vary with an average of approxi-
ume, and viscosity. The volume at the injection site is generally lower mately 34 °C, depending on the administration site.154 In fact, a tem-
and can be responsible for differences between the in vivo and the in perature of 37 °C has been applied in every setup presented in
vitro performance. This has been observed for in situ implant formu- Table 1. Otte et al. specifically examined the influence of temperature
lation, where degradation and drug release depend on the viscosity within a range of 35-37 °C on the in vitro release from naltrexone
of the matrix material.150 Testing at lower fluid volumes to accurately PLGA microparticles with a significant impact on the release behav-
mimic the SC environment leads to incomplete dissolution, especially ior.128 At 37 °C, the release was considerably higher as compared to
when poorly soluble drugs are tested. Also, the diffusion of the com- the lower temperatures. This finding is strongly related to the poly-
pound into blood circulation is not sufficiently reflected. On one mer properties and has been observed for many other PLGA and PLA
hand, the use of higher fluid volumes and surfactants can improve formulations. This is even more important when temperature
and accelerate the discrimination between drug formulations. On the changes are used to accelerate the release.132TagedEn
other hand, a change in hydrodynamics or medium composition may
alter the wetting, swelling, or dispersion properties of the drug for- TagedH2Instrumentation and Test MethodsTagedEn
mulation. Therefore, the selection of the medium is widely based on
the mechanism of release. While the release from microcrystal for- TagedPAfter a long chain of discoveries, in 1970, the first dissolution
mulations is often driven by the poor aqueous solubility of the drug apparatus was adopted by the USP.155 It was a reflection of the most
in the SC tissue,58 other formulations such as oily liquids or disper- recent efforts to provide a reproducible test system reflecting at least
sions may require their inherent structure to control the release in some aspects of human physiology and, in particular, the human gas-
the in vivo setting.151 Under these conditions, the effect of the release trointestinal tract. At present, there are no common standards for the
medium on stability properties in vitro should be taken into consider- testing of SC drug formulations. This leads to a certain diversity in the
ation. In this context, establishing IVIVCs for drug products exhibiting instrumentation and test conditions applied to these products. In the
similar formulation characteristics may support assay development.TagedEn following, we will provide an overview of some of the most relevant
TagedPThe viscosity of SC injectables has been associated with an methods. A selection of release studies including small-molecular
increasing pain sensation during the administration of the drug.152 and biotechnological compounds is presented in Table 3.TagedEn
Surprisingly, high viscosity injections (up to 15−20 cP) were better
tolerated as compared to injectables with lower viscosity. Also, the TagedPShake-Flask SetupsTagedEn
high drug and stabilizer concentrations required for the administra- TagedPA very simple but popular setup uses tubes or vials with a defined
tion of depot formulations often lead to more viscous solutions. Con- medium volume to evaluate the release. The vial is placed in shaking
sequently, the viscosities of the formulation and the SC fluid likely water baths or incubators. In an attempt to simulate the microenvi-
have a strong impact on drug diffusion as well.43 While most release ronment and hydrodynamics of the injection site, low agitation rates
media are composed of buffer systems with low viscosity, this may of 30 rpm,122,128 80 rpm117 or 100 rpm,120,129 medium volumes of up
be an often-underestimated influence.TagedEn to 50 ml122 are selected. Some methods even further reduce the

TagedEnTable 3
Selection of release studies including the drug molecules, medium, and release set-up used for the assay.

Drug Medium Release setup Refs.


120
Cytochrome c Phosphate buffer, 10 mM (pH 7.4) Shake-flask setup
127
Carbonic anhydrase / bovine serum albumin PBS (pH 7.4) Shake-flask setup with a dialysis bag
116
Monoclonal antibody formulations Physiological, carbonate-based buffer solu- SCISSOR system
tion (pH 7.4) and SCISSOR hydrogel
cartridge
58
Medroxyprogesterone acetate SSIF Dispersion releaser
33
Tacrolimus SSIF Dispersion releaser
155
Risperidone PBS (pH 7.4) USP apparatus IV
113
Naltrexone HBSS (pH 7.4) Customized capillary cell device / modified
flow-through cells
156
Flurbiprofen / Lidocaine / Risperidone Hydrogel (agarose) in PBS (pH 7.4) Hydrogel in a shake-flask setup
Interferon ß / Trypsinogen Hydrogel (agarose) Hydrogel assay in an IVISÒ system 43

117
Recombinant human insulin Hydrogel (agarose) UV imaging system
HBSS = Hanks’ balanced salts solution; IVIS = In vivo imaging system; PBS = Phosphate buffered saline; SCISSOR = Subcutaneous Injection Site Simulator; SSIF = Simulated subcutane-
ous interstitial fluid.
TagedEn TagedFiur
TagedEn1500 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

Figure 3. Illustration of the dialysis bag method used for the separation of dispersed drug formulations from the monomolecular drug. The illustrations were created with BioRen-
der (www.biorender.com).TagedEn

agedEnTPvolume to 300 ml.158 In this example, To reduce the aggregation, TagedEnProute of administration and will therefore not be discussed in more
agglomeration, and precipitation of the particles, surfactants, and sol- detail. However, interested readers are referred to a recent stimuli
ubilizers are sometimes added (e.g. sodium dodecyl sulfate, polysor- article published by the USP expert panel on New Advancements on
bate). For small volumes, the sampling volume is commonly replaced In-vitro Product Performance Testing.160 It describes the challenges
with fresh medium.117,129 Overall, this “shake-flask” technique can associated with a time-resolved separation of fine particles from the
be beneficial when compared to setups in complete stasis. Notewor- liquid.TagedEn
thy, the sensitivity of the method to fluctuations in the release profile
is widely controlled by the stirring rate. Also, further separation steps TagedPSC Injection Site Simulator (SCISSOR)TagedEn. TagedPAn interesting alternative is the
such as centrifugation or syringe filtration are often involved after SC Injection Site Simulator (“SCISSOR”) presented in Fig. 4.114 It com-
the sample collection. On one hand, the shake-flask method is highly prises an injection cartridge immersed in a container filled with a
flexible and enables the testing at various stirring rates and fluid vol- biorelevant medium. The dialysis-based cartridge is filled with hya-
umes. On the other hand, the poor standardization, of this set-up luronic acid to mimic the influence of the ECM on SC drug formula-
with regard to the size and agitation rate of magnetic stirrers and the tions. This design allows the simulation of release and diffusion
dimensions of the dissolution vessel may lead to more variability. through the ECM, as well as the uptake through the blood capillaries
Also, evaporation has to be considered. However, their clinical rele- and/or the lymph vessels.TagedEn
vance will mostly depend on the mechanism of release of the formu- TagedPThe system provides a simulated environment controlling tem-
lation.TagedEn perature, pH, and ionic strength. The interstitial pressure is simulated
by the donor cartridge due to the elastic deformation of the gel upon
TagedPDialysis Membrane-Based SetupsTagedEn injection. By increasing the fluid volume in the acceptor chamber,
TagedPFor comparison, when using dialysis-based techniques the mono- sink conditions can be adjusted. The system is usually maintained at
molecular drug is continuously separated from the release medium 34 °C and stabilized at pH 7.4 during measurements.114 To monitor
with the dialysis membrane forming a diffusion barrier. Depending the current status of the injection chamber as well as injection events
on the exact hydrodynamics of the system, the membrane can some- with impact on the stability of the drug or the excipients a micro pH
times limit the release rate. Therefore, the sensitivity of the assay probe, a thermometer, and a spectrophotometer are included. The
involves an evaluation of the membrane permeation rate. In the area spectrophotometer measures the transmittance in the injection car-
of SC drug products, two different approaches are commonly found tridge in real-time. Processes with impact on the UV/VIS spectrum
in the literature. The membrane, sometimes in combination with can be detected including the dispersion of particles as well as the
other barriers, is considered as a replacement for the physiological disintegration of the dosage form.161 Drug release can be determined
diffusion barrier posed by the SC tissue. It is used to simulate tissue by sampling from the buffer system at selected time points.TagedEn
retention and delays the availability of the drug in the acceptor TagedPThe SCISSOR system has been used for characterizing
medium (Fig. 3).TagedEn biopharmaceuticals114,116 and depot formulations of small mole-
TagedPThe operator controls the diffusion rate by selecting different cules162. Bown et al. studied the in vitro performance of a set of
membrane pore sizes, the fluid volume, temperature, and agitation monoclonal antibodies and obtained distinct profiles using the in
rate. Additionally, the liquid volume in the dialysis bag has a certain vitro setup.116 This investigation provided some evidence for the
influence on the release. It sometimes forms a static water layer that capability of the system to predict the in vivo fate of SC injectables. A
further delays the release. On one hand, the methodology provides recent study on diclofenac containing intra-articular injectables
hydrodynamics comparable to the SC tissue with a static liquid film highlighted the potential of the SCISSOR system for in vitro testing
surrounding the formulation. On the other hand, an extremely slow and comparison of the release behavior of in situ depot formula-
and variable release are major characteristics of the method.TagedEn tions.162 Major advantage of the SCISSOR is the simplified model of
TagedPAs an alternative, the membrane can be used as an analytical tool the human ECM, limiting the availability of volume fluid and the
to separate the monomolecular drug from the medium. The influence medium composition.161 However, it has limited sensitivity for com-
on release kinetics is considered an analytical error source and addi- pound interactions. One disadvantage is the limited sensitivity of the
tional measures are required to validate the assay. Also, mathemati- turbidity sensor. While macroscopic events (e.g., the disintegration of
cal models have been established to estimate the release of the drug the dosage form or dispersion of the particles) can be observed in the
from the formulation.159TagedEn UV/VIS spectrum, the background signal of the simulated ECM does
TagedPAs with other separation methods, the separation of the released not allow the analysis of specific interactions of compounds with the
drug from the medium by dialysis comes with specific advantages microenvironment. Further evaluation of this method may be
and disadvantages. These considerations are not specific to the SC required before it can be recommended for broader application.161TagedEn
TagedEn TagedFiur TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1501

Figure 4. Schematic (left) and photograph (right) of a SCISSOR system. The image was kindly provided by Karen Huffman, Pion Inc, Billerica, USA.TagedEn

TagedPDispersion ReleaserTagedEn. TagedPThe dispersion releaser (DR) is a dialysis-based TagedEnPboth compartments and reduces the impact of membrane permeabil-
device and was developed at Goethe University in 2013164. The sys- ity on the release.165 The DR has been used for the characterization of
tem is mounted into a USP dissolution apparatus I/II equipped with SC depot formulations in presence of biorelevant media such as the
standard-vessel or mini-vessel configuration.164TagedEn SSIF.33 A more recent investigation on long-acting tacrolimus micro-
TagedPThe setup comprises a donor compartment surrounded by a dialy- spheres resulted in a good prediction in vivo fate of the microspheres.
sis membrane, holding the dosage forms in the center position. The In the context of the release mechanisms discussed previously, the
dissolution vessel represents the acceptor compartment. The donor DR enables accurate detection of drug dissolution or release without
and acceptor chamber are both agitated by a paddle stirrer (Fig. 5). taking drug diffusion (through the ECM) into consideration. For the
Overall, this design facilitates rapid and controlled membrane trans- drug tacrolimus, this diffusion step was simulated in silico.33 These
port, leading to highly reproducible release profiles. Additionally, the combinations of in vitro and in silico methods will become more com-
molecular weight cut-off of the membrane can be optimized for mon in the future.TagedEn
every release experiment. Evidently, the DR does not mimic the
hydrodynamics in the SC tissue. Still, it allows a highly selective sepa- TagedPContinuous Flow-Through SetupsTagedEn
ration of different particle populations as well as sensitive detection TagedPThe flow-through cell described by the USP (dissolution apparatus
of release processes. It accelerates the medium exchange between IV) has been applied to the testing of a wide variety of parenteral
TagedEn TagedFiur

Figure 5. Schematic (left) and photograph (right) of the Pharma Test Dispersion Releaser. The images were kindly provided by Dirk Beilke, Pharma Test Apparatebau AG, Hainburg,
Germany.TagedEn
TagedEn TagedFiur
TagedEn1502 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

Figure 6. Flow-through cell setup described by the USP and one individual cell. The images were kindly provided by Michel Magnier, Sotax AG, Aesch, Switzerland.TagedEn

agedEnTPdrug formulations.156,165 It offers a wide range of commercially avail- agedEnTPapproximately 1.06 ml/min.132 However, the robustness of the sys-
able flow-through cells and a flexible reservoir volume. Hence, USP tem is often an issue. Filter membranes are affected by particles pres-
apparatus IV enables the testing of formulations where the release is ent in the formulation and lead to variable flow rates between the
either limited by drug solubility or the detection limit.165 This allows cells. Similarly, polymer migration limits the use of the continuous
the development of IVIVCs for poorly soluble drugs.166 The system flow-through setup to a certain degree.112TagedEn
comprises a pump, flow-through cells, a water bath, and medium res-
ervoirs. It can be operated with an open or a closed loop. Flow rates TagedPHydrogel-Based SetupsTagedEn
and temperatures can be adjusted by the operator.167 Dissolution TagedPAn increasing number of in vitro studies for injectables use hydro-
apparatus IV was initially designed for in vitro dissolution testing of gels as the release medium to mimic the SC tissue.43,115,124,157,168−170
controlled-release oral powders, granules, and solid dispersions The formulations are entrapped into the hydrogel matrix represent-
(Fig. 6).165TagedEn ing the SC tissue. This environment controls the fluid volume
TagedPMore often, adjustments are made to enable the testing of inject- depending on the osmotic pressure of the formulation and tempera-
ables. For example, glass beads were added to a microsphere formu- ture. Also, the hydrogel serves as a diffusion barrier.115,157,171 The
lation to avoid agglomeration in the release cell. Also, a filter can be released drugs can be quantified using UV imaging or chemical quan-
installed to prevent both glass beads and microspheres from entering tification methods. Agarose gels are most commonly used, due to the
into the medium reservoir.156,165 Compared to USP apparatuses I and ease of fabrication as well as their mechanical stability.115 Gietz et al.
II, advantages may arise from reduced agglomeration due to the described a release model based on an agarose hydrogel, simulating
higher liquid volume. Further, a potential violation of sink conditions the SC injection site. After injection of the formulation, PBS served as
can be avoided. Sampling procedures are comparable to other disso- the acceptor fluid.168 The released drug diffused through the agarose
lution methods and come with the same advantages and disadvan- gel into the liquid phase. Sink conditions were maintained by replen-
tages. Burgess and co-workers developed a dissolution test based on ishing the medium at defined time points. Another study used the in
the flow-through cell using a dialysis membrane for the separation of vivo imaging system IVISÒ to measure the fluorescence emitted by
the drug from the carrier material (Fig. 6).6 The setup is comparable fluorescence-labeled interferon released from a solid implant in mice
to other dialysis-bag methods and comes with the same limita- and a hydrogel assay. The system is designed for analyzing the biodis-
tions.163 However, the position of the dialysis adapter in the dissolu- tribution in animals and enables localization of fluorescent molecules
tion cell provides a certain standardization. Thus, the concept of (Fig. 7).43TagedEn
dissolution apparatus IV provides a good starting point for the devel- TagedPThe hydrogel assay was used to characterize the in vitro behavior
opment of SC depot formulations. Continuous perfusion of tissues as of several parenteral formulations comprising either hirudin or insu-
well as certain control of the release parameters such as flow rate lin. Leung et al. further explored the applications of the assay and
and temperature are the considerable advantages. Previous reports investigated the release of compounds with different molecular
made assumptions on the liquid flow in vivo, identifying a rate of weights.124 A UV-Vis spectrometer was used to measure the real-
TagedEn TagedFiur

Figure 7. Illustration of an agarose-based hydrogel assay using the IVISÒ system for measuring the drug release from solid implants. After the implantation of the solid implant into
animals and the hydrogel, the fluorescence was monitored over time. The illustration was created with BioRender (www.biorender.com).TagedEn
TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1503

TagedEnPtime drug concentration in the buffer system. Biorelevance was dem- TagedEnPpreclinical models may not be suitable to predict absorption from the
onstrated by means of IVIVC.TagedEn SC tissue. However, the size cut-off of 100 nm was also consistent
with porcine models suggesting that these animals may provide suffi-
TagedPQuantification MethodsTagedEn cient information to study the burst effect related to lymphatic
TagedPMost performance assays can be combined with various quantifi- uptake.16TagedEn
cation methods. Imaging methods have certain advantages. They TagedPDepot formulations for SC administration are commonly adminis-
enable the observation of release processes in vitro and in vivo.43TagedEn tered into a small volume present at the injection site. The protein
TagedPThis includes the stability of the injected components, swelling, concentration in the interstitial space is lower as compared to the
and erosion behavior of formulations, and drug transport. Impor- blood plasma.143 Still, these proteins can have a stabilizing effect on
tantly, every quantification method comes with its weaknesses and particle dispersions.58 Since agglomeration often reduces the fine
strengths. While conventional high-performance liquid chromatog- particle fraction and proteins counteract this process, the in vitro
raphy setups detect the drug from various matrices, in situ methods studies should include physical stability in the presence and absence
enable a continuous detection of more time points. These differences of serum proteins.58 Currently, SSIF is the only release medium
are not specific to the SC route of administration and will therefore including this essential component.33,58,144TagedEn
not be discussed in more detail. With regards to imaging techniques, TagedPAmong other things, drug dissolution is affected by the fluid vol-
diagnostic ultrasound imaging, fluorescence imaging, and UV-Vis ume, temperature, aqueous solubility of the compound, and the par-
imaging are amongst the most popular non-destructive analytical titioning into lipophilic tissues. While temperature usually varies in a
methods.122 One of the commercial set-ups that have been used for range of 3-5 °C and often remains without considerable impact on
the testing of drug release is the IVISÒ . The in vivo scanner was pharmacokinetics, the fluid volume in the SC tissue is flexible and
designed for the analysis of biodistributions in small animals such as depends on the osmotic pressure of the formulation. However, with
rodents or mice (Fig. 7). After illumination of the inner chamber with regard to dissolution, aqueous solubility and particle size remain the
the light of a specific wavelength, the fluorescence emission from the most critical parameters. While temperature has only a minor influ-
sample is detected resulting in a two-dimensional and, in the latest ence on the solubility of microcrystal or nanocrystal formulations, it
versions of the instrument, three-dimensional image. After implanta- sometimes represents an important trigger for the release from tem-
tion into a hydrogel, the IVISÒ system has been used to detect the perature-sensitive polymer128 or lipid carriers.1 Therefore, whenever
release of a fluorescence-labeled protein from solid implant the material properties indicate such a relationship, the temperature
formulations.43TagedEn should be monitored in the assay system over a wide temperature
TagedPAt present, there are several methods under investigation, to con- range to ensure that there are no unexpected changes in the release
tinuously monitor the drug release. While instruments such as the kinetics observed. For poorly soluble compounds, partitioning into
SCISSOR or UV imaging methods172 are more suitable for the detec- the SC fat layer affects bioavailability and dissolution behavior as
tion in a controlled in vitro environment, fluorescence imaging, near- well.99 To study these interactions a biphasic release medium can be
infra-red, or magnetic resonance imaging are suitable to detect used. Unfortunately, animal studies in mice are of limited value in
changes in the performance in vitro and in vivo.173,174TagedEn the prediction of these effects. The composition of the SC tissue dif-
fers from the human situation. For studies investigating drug distri-
TagedH1Authors’ RecommendationsTagedEn bution in the skin layers, rats or non-human primates are of higher
value.33,46TagedEn
TagedPTo develop in vitro assays capable of predicting the in vivo TagedPWith a growing number of biotechnological compounds entering
performance, the mechanisms involved in the release from the drug the global healthcare market, metabolic stability is becoming more
product must be reflected by method and medium. Various influen- important. For insoluble compounds with poor metabolic stability,
ces on drug absorption were summarized in the previous sections. chemical degradation may accelerate drug dissolution. The degrada-
Together with a dedicated investigation of the formulation properties tion step reduces the concentration of the free compound resulting in
and responses, they are the basis for the selection of the release more dissolution. Under these conditions, degradability is an impor-
conditions.TagedEn tant release mechanism and requires an investigation in vitro and in
TagedPOne of the critical influences on drug absorption is the amount of vivo. For example, hydrolysis of tacrolimus is not affected by the pres-
the unbound drug present in the dosage form. Depending on the ence of serum33 while metabolic stability of other drugs depends on
exact manufacturing process, liquid dispersions have a higher risk for the presence of enzymes and requires more effort in the simula-
safety-relevant changes in the encapsulation over time. Initial disper- tion.135 To predict and investigate the effects of the formulation on
sion of this weakly bound fraction leads to “burst release” with a high the chemical stability of the compound in vivo, the preclinical model
probability of reaching toxicologically relevant concentrations in the plays an important role. Enzyme expression levels between different
blood plasma. In the in vivo setting, this “burst effect” often overlaps species may vary and should therefore be considered when develop-
with the initial distribution of the compound after injection and can ing the drug formulations. With regard to the development of perfor-
be hard to distinguish from the migration of nanosized particles into mance assays, the expected degradation mechanisms can be
the lymphatic capillaries. Therefore, both parameters, the fine parti- simulated by addition of enzymes to the release medium. Knowing
cle fraction and the release behavior should be accurately moni- the processes affecting the chemical stability of the compound is,
tored.33 Both methods together, release testing and physical stability therefore, is of utmost importance.TagedEn
testing, are potentially sensitive enough to detect these safety issues. TagedPFor many compounds with moderate or high aqueous solubility,
Noteworthy, the release assays utilized in the detection of “burst other processes such as the diffusion of the drug to the nearest blood
effects” must be sensitive for rapid fluctuations in the release rate.162 vessel or the unspecific interactions between the drug and the carrier
Conventional dialysis approaches or hydrogels may not be able to have a stronger impact on bioavailability. The differences in drug
sufficiently discriminate these changes. The inherent barrier proper- absorption following SC and intramuscular injection are more pro-
ties of the assay delay the release and reduce reproducibility. More nounced for biologics175 than for poorly soluble compounds.176 Diffu-
rapid separation techniques, even when using non-physiological sion becomes the rate-limiting step in the absorption process. Here,
hydrodynamics and fluid volume, are more appropriate. In the pre- assay designs measuring this diffusion rate often provide a better
clinical setting, most studies investigating the lymphatic transport of estimate of the drug absorption.43 The molecular size and protein
drugs into systemic circulation are carried out in rodents. These binding of a drug becomes a more important parameter. In this
TagedEn1504 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

TagedEnPcontext, breaking down the absorption into the individual processes TagedEnPfact, the perfect balance between sensitivity, selectivity, and biorelev-
and simulating these steps either in vitro or in silico will enable a bet- ance commonly leads to the best outcomes.TagedEn
ter prediction than dissolution testing alone.TagedEn
TagedPMore advanced formulation designs control the release by using a TagedH1Declaration of InterestsTagedEn
specific combination of excipients. With these ingredients controlling
the release behavior, their interaction with the physiological micro- TagedPThe authors declare that they have no known competing financial
environment becomes more important. For example, changes in the interest.TagedEn
physiological fluid volume at the injection site over time may affect
the release behavior from in situ implant formulations.151 Consider-
ing that the influx of liquid varies over time, the release processes TagedH1AcknowledgmentsTagedEn
would hardly be reflected by an in vitro setup with a static fluid vol-
ume. Hydrogel systems provide a flexible response to dissolution, TagedPThe authors acknowledge Jennifer Dressman for her years of dedi-
swelling, and osmotic pressure of the formulation and could poten- cation to biorelevant release testing, her strong scientific and per-
tially lead to better outcomes.43 More than a representation of the sonal support, and for treating her staff, students, and co-workers as
physiological environment, this biopredictive assay activates a physi- a second family.TagedEn
ologically relevant trigger. Similarly, for some formulations, affinities TagedPD.L. acknowledges the National University of Singapore (NUS) and
between the molecules in the formulation can be weakened by A*STAR for providing him with a scholarship in the NUS Pharmacy
endogenous binding partners. For liposomes, the exchange of phos- graduate research program. Also, all authors acknowledge Pion Inc.
pholipids between the vesicle and biomembranes has been observed (Billerica, USA), Pharma Test Apparatebau AG (Hainburg, Germany),
and potentially affects membrane permeability. For the SC formula- and Sotax AG (Aesch, Switzerland) for providing us with illustration
tion InsulatardÒ , a complex of insulin, protamine, and zinc, insulin is material as well as NUS and the Ministry of Education of Singapore
continuously replaced by negatively charged binding partners such for financial support (grant no. R-148-000-282-133 and R-148-000-
as heparin or chondroitin.177 Under these conditions, a release assay 297-114).TagedEn
not providing suitable binding partners may not be able to forecast
the in vivo performance.TagedEn TagedH1ReferencesTagedEn
TagedPAs outlined by the previous sections, the biocompatibility of the
carrier is a key issue in its performance. Morphological changes of TagedP 1. Rahnfeld L, Luciani P. Injectable lipid-based depot formulations: where do we
stand? Pharmaceutics. 2020;12(6). https://doi.org/10.3390/pharmaceu-
the physiological environment can be a gamechanger in the release
tics12060567.TagedEn
and diffusion processes and make predictions more challenging. TagedP 2. Kwatra S, Taneja G, Nasa N. Alternative routes of drug administration - transder-
With formulations remaining at the injection site over a considerable mal, pulmonary & parenteral. Indo Glob J Pharm Sci. 2012;2(4):409–426.TagedEn
time, the risk for long-term effects, inflammation, or immune TagedP 3. Sugano K, Okazaki A, Sugimoto S, Tavornvipas S, Omura A, Mano T. Solubility and
dissolution profile assessment in drug discovery. Drug Metab Pharmacokinet.
responses is higher as compared to other dosage forms. These 2007;22(4):225–254. https://doi.org/10.2133/dmpk.22.225.TagedEn
responses are not limited to the drug or excipients but may occur TagedP 4. Schmier J, Ogden K, Nickman N, et al. Costs of providing infusion therapy for
due to the trauma caused by the injection. Also, the disassembly of rheumatoid arthritis in a hospital-based infusion center setting. Clin Ther.
2017;39(8):1600–1617. https://doi.org/10.1016/j.clinthera.2017.06.007.TagedEn
the formulation at the injection site may lead to inflammation reac- TagedP 5. Turner MR, Balu-Iyer SV. Challenges and opportunities for the subcutaneous
tions.178 At present, the prediction of inflammation or immune reac- delivery of therapeutic proteins. J Pharm Sci. 2018;107(5):1247–1260. https://doi.
tions still requires a significant number of preclinical studies. The org/10.1016/j.xphs.2018.01.007.TagedEn
TagedP 6. Bhardwaj U, Burgess DJ. A novel USP apparatus 4 based release testing method for
implications and models in vitro and in vivo have been discussed in dispersed systems. Int J Pharm. 2010;388(1-2):287–294. https://doi.org/10.1016/j.
more detail by Jarvi and Balu-Iyer.99 Importantly, these responses are ijpharm.2010.01.009.TagedEn
difficult to predict using a release assay and are not within the scope TagedP 7. Collins DS, Sanchez-Fe lix M, Badkar AV, Mrsny R. Accelerating the development
of novel technologies and tools for the subcutaneous delivery of biotherapeutics.
of this review article.TagedEn J Control Release. 2020;321:475–482. https://doi.org/10.1016/j.jconrel.2020.02.
TagedPOur recommendations provide some advice to the operator in the 036.TagedEn
selection of the in vitro conditions during the development of perfor- TagedP 8. Yoo J, Won Y-Y. Phenomenology of the Initial burst release of drugs from PLGA
microparticles. ACS Biomater Sci Eng. 2020;6(11):6053–6062. https://doi.org/
mance assays. However, the abovementioned findings were the
10.1021/acsbiomaterials.0c01228.TagedEn
result of a thorough understanding of the mechanisms of release. TagedP 9. Lee C, Guo H, Klinngam W, et al. Berunda polypeptides: biheaded rapamycin car-
We, therefore, caution the definition of general standards. The formu- riers for subcutaneous treatment of autoimmune dry eye disease. Mol Pharm.
lation design must be reflected by the assay to a certain extent.TagedEn 2019;16(7):3024–3039. https://doi.org/10.1021/acs.molpharmaceut.9b00263.TagedEn
TagedP 10. Sheikh Hassan A, Sapin A, Lamprecht A, Emond E, El Ghazouani F, Maincent
P. Composite microparticles with in vivo reduction of the burst release
TagedH1ConclusionTagedEn effect. Eur J Pharm Biopharm. 2009;73(3):337–344. https://doi.org/10.1016/j.
ejpb.2009.07.009.TagedEn
TagedP 11. Ni Q, Chen W, Tong L, Cao J, Ji C. Preparation of novel biodegradable ropivacaine
agedPIT n conclusion, the SC route of administration provides an effective microspheres and evaluation of their efficacy in sciatic nerve block in mice. Drug
delivery platform for many compounds by improving their bioavail- Des Devel Ther. 2016;10:2499–2506. https://doi.org/10.2147/dddt.S110742).TagedEn
ability or sustaining the release over a considerable period. Still, a TagedP 12. Robinson NB, Krieger K, Khan FM, et al. The current state of animal models in
research: a review. Int J Surg. 2019;72:9–13. https://doi.org/10.1016/j.
number of overlapping processes are involved in the absorption of ijsu.2019.10.015.TagedEn
the drug, including lymphatic uptake, the release of the drug from TagedP 13. Pound P, Ritskes-Hoitinga M. Is it possible to overcome issues of external validity
the delivery system, post-injection agglomeration, or inflammatory in preclinical animal research? Why most animal models are bound to fail. J
Transl Med. 2018;16. https://doi.org/10.1186/s12967-018-1678-1.TagedEn
responses. Knowledge gaps affect the predictive power of the assays TagedP 14. Wu Z, Hassan D, Shaw JP. In-vitro prediction of bioavailability following extravas-
and still require a thorough investigation of the most important influ- cular injection of poorly soluble drugs: an insight into clinical failure and the role
ences.TagedEn of delivery systems. J Pharm Pharmacol. 2013;65(10):1429–1439. https://doi.org/
10.1111/jphp.12114.TagedEn
TagedPBy gaining a better understanding of the mechanisms of release,
TagedP 15. Fenner J, Clark RAF. Chapter 1 - Anatomy, physiology, histology, and immunohis-
predictability of absorption can be improved and its impact on the tochemistry of human skin. In: Albanna MZ, Holmes IV JH, eds. Skin Tissue Engi-
plasma drug concentration better simulated. This ensures that novel neering and Regenerative Medicine. 1st ed. Boston: Academic Press; 2016:1–17.
drug formulations are safe and effective when entering clinical trials. https://doi.org/10.1016/B978-0-12-801654-1.00001-2.TagedEn
TagedP 16. McLennan DN, Porter CJH, Charman SA. Subcutaneous drug delivery and the role
There is a common misunderstanding in dissolution science that a of the lymphatics. Drug Disocov Today. 2005;2(1):89–96. https://doi.org/10.1016/
more physiological assay automatically leads to better predictions. In j.ddtec.2005.05.006.TagedEn
TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1505

TagedP 17. Prettyman J. Subcutaneous or intramuscular? Confronting a parenteral adminis- TagedEnPtechniques. J Control Release. 2016;235:352–364. https://doi.org/10.1016/j.jcon-
tration dilemma. Medsurg Nurs. 2005;14(2):93–99.TagedEn rel.2016.06.013.TagedEn
TagedP 18. Rodriguez-Aller M, Guillarme D, Veuthey J-L, Gurny R. Strategies for formulating TagedP 44. Jucker BM, Fuchs EJ, Lee S, et al. Multiparametric magnetic resonance imaging to
and delivering poorly water-soluble drugs. J Drug Deliv Sci Technol. 2015;30:342– characterize cabotegravir long-acting formulation depot kinetics in healthy adult
351. https://doi.org/10.1016/j.jddst.2015.05.009.TagedEn volunteers. Br J Clin Pharmacol. 2021. https://doi.org/10.1111/bcp.14977.TagedEn
TagedP 19. Sigfridsson K, Lundqvist A, Strimfors M. Subcutaneous administration of nano- TagedP 45. McLennan DN, Porter CJH, Edwards GA, Heatherington AC, Martin SW, Charman
and microsuspensions of poorly soluble compounds to rats. Drug Dev Ind Pharm. SA. The absorption of darbepoetin alfa occurs predominantly via the lymphatics
2014;40(4):511–518. https://doi.org/10.3109/03639045.2013.771645.TagedEn following subcutaneous administration to sheep. Pharm Res. 2006;23(9):2060–
TagedP 20. Blume G, Cevc G. Liposomes for the sustained drug release in vivo. Biochim Bio- 2066. https://doi.org/10.1007/s11095-006-9064-8.TagedEn
phys Acta. 1990;1029:91–97. https://doi.org/10.1016/0005-2736(90)90440-Y.TagedEn TagedP 46. Datta-Mannan A, Estwick S, Zhou C, et al. Influence of physiochemical properties
TagedP 21. Kaminskas LM, Kota J, McLeod VM, Kelly BD, Karellas P, Porter CJ. PEGylation of on the subcutaneous absorption and bioavailability of monoclonal antibodies.
polylysine dendrimers improves absorption and lymphatic targeting following mAbs. 2020;12(1). https://doi.org/10.1080/19420862.2020.1770028. 1770028-
SC administration in rats. J Control Release. 2009;140(2):108–116. https://doi.org/ 1770028.TagedEn
10.1016/j.jconrel.2009.08.005.TagedEn TagedP 47. Chandran PL, Horkay F. Aggrecan, an unusual polyelectrolyte: review of solution
TagedP 22. Mu € ller RH, Maassen S, Weyhers H, Mehnert W. Phagocytic uptake and cytotoxic- behavior and physiological implications. Acta Biomater. 2012;8(1):3–12. https://
ity of solid lipid nanoparticles (SLN) sterically stabilized with poloxamine 908 doi.org/10.1016/j.actbio.2011.08.011.TagedEn
and poloxamer 407. J Drug Target. 1996;4(3):161–170. https://doi.org/10.3109/ TagedP 48. Lapcík Jr. L, Lapcík L, De Smedt S, Demeester J, Chabrecek P. Hyaluronan: prepara-
10611869609015973.TagedEn tion, structure, properties, and applications. Chem Rev. 1998;98(8):2663–2684.
TagedP 23. Allen TM, Hansen C, Rutledge J. Liposomes with prolonged circulation times: fac- https://doi.org/10.1021/cr941199z.TagedEn
tors affecting uptake by reticuloendothelial and other tissues. Biochim Biophys TagedP 49. Collins DS, Kourtis LC, Thyagarajapuram NR, et al. Optimizing the bioavailability
Acta. 1989;981(1):27–35. https://doi.org/10.1016/0005-2736(89)90078-3.TagedEn of subcutaneously administered biotherapeutics through mechanochemical driv-
TagedP 24. Kim YH, Min KH, Wang Z, et al. Development of sialic acid-coated nanoparticles ers. Pharm Res. 2017;34(10):2000–2011. https://doi.org/10.1007/s11095-017-
for targeting cancer and efficient evasion of the immune system. Theranostics. 2229-9.TagedEn
2017;7(4):962–973. https://doi.org/10.7150/thno.19061.TagedEn TagedP 50. Yadav S, Liu J, Shire SJ, Kalonia DS. Specific interactions in high concentration
TagedP 25. Hawley AE, Illum L, Davis SS. Lymph node localisation of biodegradable antibody solutions resulting in high viscosity. J Pharm Sci. 2010;99(3):1152–
nanospheres surface modified with poloxamer and poloxamine block co-pol- 1168. https://doi.org/10.1002/jps.21898.TagedEn
ymers. FEBS Lett. 1997;400(3):319–323. https://doi.org/10.1016/s0014-5793 TagedP 51. Mach H, Gregory SM, Mackiewicz A, et al. Electrostatic interactions of monoclonal
(96)01408-1.TagedEn antibodies with subcutaneous tissue. Ther Deliv. 2011;2(6):727–736. https://doi.
TagedP 26. Hawley AE, Illum L, Davis SS. The effect of lymphatic oedema on the uptake of col- org/10.4155/tde.11.31.TagedEn
loids to the lymph nodes. Biopharm Drug Dispos. 1998;19(3):193–197. https://doi. TagedP 52. Li ST, Katz EP. An electrostatic model for collagen fibrils. The interaction of recon-
org/10.1002/(sici)1099-081x(199804)19:3<193::aid-bdd88>3.0.co;2-v.TagedEn stituted collagen with Ca++, Na+, and Cl. Biopolymers. 1976;15(8):1439–1460.
TagedP 27. Oussoren C, Zuidema J, Crommelin DJA, Storm G. Lymphatic uptake and biodistri- https://doi.org/10.1002/bip.1976.360150802.TagedEn
bution of liposomes after subcutaneous injection. II. Influence of liposomal size, TagedP 53. Uchisawa H, Okuzaki B-i, Ichita J, Matsue H. Binding between calcium ions
lipid compostion and lipid dose. Biochim Biophys Acta. 1997;1328(2):261–272. and chondroitin sulfate chains of salmon nasal cartilage glycosaminoglycan.
https://doi.org/10.1016/s0005-2736(97)00122-3.TagedEn Int Congress Ser. 2001;1223:205–220. https://doi.org/10.1016/S0531-5131
TagedP 28. Swartz M. The physiology of the lymphatic system. Adv Drug Deliv Rev. 2001;50 (01)00458-7.TagedEn
(1-2):3–20. https://doi.org/10.1016/s0169-409x(01)00150-8.TagedEn TagedP 54. Fernqvist E, Linde B, Ostman € J, Gunnarsson R. Effects of physical exercise on insu-
TagedP 29. Oussoren C, Storm G. Liposomes to target the lymphatics by subcutaneous lin absorption in insulin-dependent diabetics. A comparison between human and
administration. Adv Drug Deliv Rev. 2001;50(1-2):143–156. https://doi.org/ porcine insulin. Clin Physiol. 1986;6(6):489–497. https://doi.org/10.1111/j.1475-
10.1016/s0169-409x(01)00154-5.TagedEn 097X.1986.tb00782.x).TagedEn
TagedP 30. Devalapally H, Chakilam A, Amiji MM. Role of nanotechnology in pharmaceutical TagedP 55. Richard JL, Rodier M, Cavalie  G, Mirouze J, Monnier L. Human (recombinant DNA)
product development. J Pharm Sci. 2007;96(10):2547–2565. https://doi.org/ and porcine NPH insulins are unequally effective in diabetic patients. A compara-
10.1002/jps.20875.TagedEn tive study using continuous blood glucose monitoring. Acta Diabetol Lat. 1984;21
TagedP 31. Allen TM, Hansen CB, Guo LS. Subcutaneous administration of liposomes: a com- (3):211–217. https://doi.org/10.1007/BF02642894.TagedEn
parison with the intravenous and intraperitoneal routes of injection. Biochim Bio- TagedP 56. Wang W, Chen N, Shen X, et al. Lymphatic transport and catabolism of therapeu-
phys Acta. 1993;1150(1):9–16. https://doi.org/10.1016/0005-2736(93)90115-g.TagedEn tic proteins after subcutaneous administration to rats and dogs. Drug Metab Dis-
TagedP 32. Oussoren C, Velinova M, Scherphof G, van der Want JJ, van Rooijen N, Storm G. pos. 2012;40(5):952–962. https://doi.org/10.1124/dmd.111.043604.TagedEn
Lymphatic uptake and biodistribution of liposomes after subcutaneous injection. TagedP 57. Zbyszynski P, Toraason I, Repp L, Kwon GS. Probing the subcutaneous absorption
IV. Fate of liposomes in regional lymph nodes. Biochim Biophys Acta. 1998;1370 of a PEGylated FUD peptide nanomedicine via in vivo fluorescence imaging. Nano
(2):259–272. https://doi.org/10.1016/s0005-2736(97)00275-7.TagedEn Convergence. 2019;6(1). https://doi.org/10.1186/s40580-019-0192-3. 22-15.TagedEn
TagedP 33. Gao GF, Ashtikar M, Kojima R, et al. Predicting drug release and degradation TagedP 58. Gao GF, Thurn M, Wendt B, Parnham MJ, Wacker MG. A sensitive in vitro perfor-
kinetics of long-acting microsphere formulations of tacrolimus for subcuta- mance assay reveals the in vivo drug release mechanisms of long-acting medrox-
neous injection. J Control Release. 2021;329. https://doi.org/10.1016/j.jcon- yprogesterone acetate microparticles. Int J Pharm. 2020;586: 119540. https://doi.
rel.2020.11.055. 372-284.TagedEn org/10.1016/j.ijpharm.2020.119540.TagedEn
TagedP 34. Trevaskis NL, Kaminskas LM, Porter CJ. From sewer to saviour - targeting the lym- TagedP 59. Zhang L, Navaratna T, Thurber GM. A helix-stabilizing linker improves sub-
phatic system to promote drug exposure and activity. Nat Rev Drug Discov. cutaneous bioavailability of a helical peptide independent of linker lipophi-
2015;14(11):781–803. https://doi.org/10.1038/nrd4608.TagedEn licity. Bioconjug Chem. 2016;27(7):1663–1672. https://doi.org/10.1021/acs.
TagedP 35. Hawley AE, Davis SS, Illum L. Targeting of colloids to lymph nodes: influence of bioconjchem.6b00209.TagedEn
lymphatic physiology and colloidal characteristics. Adv Drug Deliv Rev. 1995;17 TagedP 60. Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (plga) as biodegradable con-
(1):129–148.TagedEn trolled drug delivery carrier. Polymers. 2011;3(3):1377–1397.TagedEn
TagedP 36. Khullar OV, Griset AP, Gibbs-Strauss SL, et al. Nanoparticle migration and delivery TagedP 61. Wissing SA, Kayser O, Mu € ller RH. Solid lipid nanoparticles for parenteral drug
of Paclitaxel to regional lymph nodes in a large animal model. J Am Coll Surg. delivery. Adv Drug Deliv Rev. 2004;56(9):1257–1272. https://doi.org/10.1016/j.
2012;214(3):328–337. https://doi.org/10.1016/j.jamcollsurg.2011.11.006.TagedEn addr.2003.12.002.TagedEn
TagedP 37. Summerfield A, Meurens F, Ricklin ME. The immunology of the porcine skin and TagedP 62. Guo Z, Bo D, He Y, Luo X, Li H. Degradation properties of chitosan microspheres/
its value as a model for human skin. Mol Immunol. 2015;66(1):14–21. https://doi. poly(L-lactic acid) composite in vitro and in vivo. Carbohydr Polym. 2018;193:1–
org/10.1016/j.molimm.2014.10.023.TagedEn 8. https://doi.org/10.1016/j.carbpol.2018.03.067.TagedEn
TagedP 38. Oussoren C, Storm G. Lymphatic uptake and biodistribution of liposomes TagedP 63. zur Mu € hlen A, Schwarz C, Mehnert W. Solid lipid nanoparticles (SLN) for con-
after subcutaneous injection: III. Influence of surface modification with poly trolled drug delivery−drug release and release mechanism. Eur J Pharm Biopharm.
(ethyleneglycol). Pharm Res. 1997;14(10):1479–1484. https://doi.org/ 1998;45(2):149–155. https://doi.org/10.1016/s0939-6411(97)00150-1.TagedEn
10.1023/a:1012145410859.TagedEn TagedP 64. Siegel SJ, Kahn JB, Metzger K, Winey KI, Werner K, Dan N. Effect of drug type on
TagedP 39. Oussoren C, Zuidema J, Crommelin DJA, Storm G. Lymphatic Uptake and Biodistri- the degradation rate of PLGA matrices. Eur J Pharm Biopharm. 2006;64(3):287–
bution of Liposomes after Subcutaneous Injection I. Influence of the Anatomical 293. https://doi.org/10.1016/j.ejpb.2006.06.009.TagedEn
Site of Injection. J Liposome Res. 1997;7(1):85–99. https://doi.org/10.3109/ TagedP 65. Xu Y, Kim C-S, Saylor DM, Koo D. Polymer degradation and drug delivery in PLGA-
08982109709035487.TagedEn based drug-polymer applications: a review of experiments and theories. J Biomed
TagedP 40. Nebendahl K. Chapter 24 - Routes of administration. In: Krinke GJ, ed. The Labora- Mater Res B. 2017;105(6):1692–1716. https://doi.org/10.1002/jbm.b.33648.TagedEn
tory Rat. London: Academic Press; 2000:463–483. https://doi.org/10.1016/B978- TagedP 66. Feczko  P, Pleli S, Denk H, Ro € del V, Wacker. Theranostic sorafenib-loaded
012426400-7.50063-7.TagedEn polymeric nanocarriers manufactured by enhanced gadolinium conjugation
TagedP 41. Harris JM, Chess RB. Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov. techniques. Pharmaceutics. 2019;11(10):489. https://doi.org/10.3390/pharma-
2003;2(3):214–221. https://doi.org/10.1038/nrd1033.TagedEn ceutics11100489.TagedEn
TagedP 42. Fishburn CS. The pharmacology of PEGylation: balancing PD with PK to generate TagedP 67. Go € pferich A. Mechanisms of polymer degradation and erosion. Biomaterials.
novel therapeutics. J Pharm Sci. 2008;97(10):4167–4183. https://doi.org/10.1002/ 1996;17(2):103–114. (https://doi.org/10.1016/0142-9612(96)85755-3).TagedEn
jps.21278.TagedEn TagedP 68. Yoon SJ, Kim SH, Ha HJ, et al. Reduction of inflammatory reaction of poly(d,l-lac-
TagedP 43. Beyer S, Xie L, Schmidt M, et al. Optimizing novel implant formulations for the tic-co-glycolic Acid) using demineralized bone particles. Tissue Eng Part A.
prolonged release of biopharmaceuticals using in vitro and in vivo imaging 2008;14(4):539–547. https://doi.org/10.1089/tea.2007.0129.TagedEn
TagedEn1506 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

TagedP 69. Anderson JM, Shive MS. Biodegradation and biocompatibility of PLA and PLGA TagedP 95. Heinemann L. Insulin absorption from lipodystrophic areas: a (neglected) source
microspheres. Adv Drug Deliv Rev. 1997;28(1):5–24. https://doi.org/10.1016/ of trouble for insulin therapy? J Diabetes Sci Technol. 2010;4(3):750–753. https://
s0169-409x(97)00048-3.TagedEn doi.org/10.1177/193229681000400332.TagedEn
TagedP 70. Grady H, Elder D, Webster GK, et al. Industry's view on using quality control, bio- TagedP 96. Ratner BD. Chapter 3 - The biocompatibility of implant materials. In: Badylak SF,
relevant, and clinically relevant dissolution tests for pharmaceutical develop- ed. Host Response to Biomaterials. Oxford: Academic Press; 2015:37–51. https://
ment, registration, and commercialization. J Pharm Sci. 2018;107(1):34–41. doi.org/10.1016/B978-0-12-800196-7.00003-7.TagedEn
https://doi.org/10.1016/j.xphs.2017.10.019.TagedEn TagedP 97. Jones K. 2015. Chapter 9 - Fibrotic response to biomaterials and all associated
TagedP 71. Ford Versypt AN, Pack DW, Braatz RD. Mathematical modeling of drug delivery sequence of fibrosis. In Badylak SF, Host Response to Biomaterials, Oxford: Aca-
from autocatalytically degradable PLGA microspheres. Rev. 2013;165(1):29–37. demic Press. p 189-237. (https://doi.org/10.1016/B978-0-12-800196-7.00009-8).TagedEn
https://doi.org/10.1016/j.jconrel.2012.10.015.TagedEn TagedP 98. Dingman R, Balu-Iyer SV. Immunogenicity of Protein Pharmaceuticals. J Pharm Sci.
TagedP 72. Gleadall A, Pan J, Kruft M-A, Kellom€aki M. Degradation mechanisms of biore- 2019;108(5):1637–1654. https://doi.org/10.1016/j.xphs.2018.12.014.TagedEn
sorbable polyesters. Part 1. Effects of random scission, end scission and auto- TagedP 99. Jarvi NL, Balu-Iyer SV. Immunogenicity challenges associated with subcutaneous
catalysis. Acta Biomater. 2014;10(5):2223–2232. https://doi.org/10.1016/j. delivery of therapeutic proteins. BioDrugs. 2021;35(2):125–146. https://doi.org/
actbio.2013.12.039.TagedEn 10.1007/s40259-020-00465-4.TagedEn
TagedP 73. Siepmann J, Elkharraz K, Siepmann F, Klose D. How autocatalysis accelerates drug TagedP100. Gradel AKJ, Porsgaard T, Lykkesfeldt J, et al. Factors affecting the absorption of
release from PLGA-based microparticles: a quantitative treatment. Biomacromole- subcutaneously administered insulin: effect on variability. J Diabetes Res.
cules. 2005;6(4):2312–2319. https://doi.org/10.1021/bm050228k.TagedEn 2018;2018:1–17. https://doi.org/10.1155/2018/1205121.TagedEn
TagedP 74. Yoo HS, Park TG. Biodegradable polymeric micelles composed of doxorubicin con- TagedP101. Moriya Y, Kogame A, Tagawa Y, et al. The enhancement of subcutaneous first-pass
jugated PLGA−PEG block copolymer. J Control Release. 2001;70:63–70. https:// metabolism causes nonlinear pharmacokinetics of TAK-448 after a single subcu-
doi.org/10.1016/s0168-3659(00)00340-0.TagedEn taneous administration to rats. Drug Metab Dispos. 2019;47(9):1004–1012.
TagedP 75. Hu Q, Prakash J, Rijcken CJF, Hennink WE, Storm G. High systemic availability of https://doi.org/10.1124/dmd.119.087148.TagedEn
core-crosslinked polymeric micelles after subcutaneous administration. Int J TagedP102. Komada F, Okumura K, Hori R. Fate of porcine and human insulin at the subcuta-
Pharm. 2016;514(1):112–120. https://doi.org/10.1016/j.ijpharm.2016.09.030.TagedEn neous injection site. Ii. In vitro degradation of insulins in the subcutaneous tissue
TagedP 76. Deadman CM, Kellaway IW, Yasin M, Dickinson PA, Murdan S. An investigation of the rat. J Pharmacobiodyn. 1985;8(1):33–40. https://doi.org/10.1248/
into the influence of drug lipophilicity on the in vivo absorption profiles from bpb1978.8.33.TagedEn
subcutaneous microspheres and in situ forming depots. J Control Release. TagedP103. Murakami T, Misaki M, Kojima Y, et al. Effect of absorption promoters on subcuta-
2007;122(1):79–85. https://doi.org/10.1016/j.jconrel.2007.06.013.TagedEn neous absorption of human epidermal growth factor in rats. J Pharm Sci. 1993;82
TagedP 77. Johnson RJ, Kerr CL, Enouri SS, Modi P, Lascelles BDX, Del Castillo JRE. Pharmaco- (3):236–239. https://doi.org/10.1002/jps.2600820303.TagedEn
kinetics of liposomal encapsulated buprenorphine suspension following subcuta- TagedP104. Esposito S, de Leonibus ML, Ingenito R, Bianchi E, Orsatti L, Monteagudo E. A liq-
neous administration to cats. J Vet Pharmacol Ther. 2017;40(3):256–269. https:// uid chromatography high-resolution mass spectrometry in vitro assay to assess
doi.org/10.1111/jvp.12357.TagedEn metabolism at the injection site of subcutaneously administered therapeutic pep-
TagedP 78. Blanco MD, Bernardo MV, Sastre RL, Olmo R, Mun ~ iz E, Teijo
n JM. Preparation of tides. J Pharm Biomed Anal. 2018;159:449–458. https://doi.org/10.1016/j.
bupivacaine-loaded poly(e-caprolactone) microspheres by spray drying: drug jpba.2018.07.009.TagedEn
release studies and biocompatibility. Eur J Pharm Biopharm. 2003;55(2):229–236. TagedP105. Iwao Y, Anraku M, Hiraike M, et al. The structural and pharmacokinetic properties
https://doi.org/10.1016/s0939-6411(02)00169-8.TagedEn of oxidized human serum albumin, advanced oxidation protein products (AOPP).
TagedP 79. Huang X, Brazel CS. On the importance and mechanisms of burst release in 2021. Drug Metab Pharmacokinet. 2006;21(2):140–146. https://doi.org/10.2133/
matrix-controlled drug delivery systems. J Control Release. 2001;73(2-3):121– dmpk.21.140.TagedEn
136. https://doi.org/10.1016/s0168-3659(01)00248-6.TagedEn TagedP106. Iwao Y, Anraku M, Yamasaki K, et al. Oxidation of Arg-410 promotes the elimina-
TagedP 80. Evans DC. Predicting Injection Site Drug Precipitation. Tucson, AZ: The University of tion of human serum albumin. Biochim. Biophys. Acta Proteins Proteom..
Arizona; 2013.TagedEn 2006;1764(4):743–749. https://doi.org/10.1016/j.bbapap.2006.01.011.TagedEn
TagedP 81. Tiemessen H, van Hoogevest P, Leigh ML. Characteristics of a novel phospholipid- TagedP107. Noga M, Edinger D, Kl€ager R, et al. The effect of molar mass and degree of
based depot injectable technology for poorly water-soluble drugs. Eur J Pharm hydroxyethylation on the controlled shielding and deshielding of hydrox-
Biopharm. 2004;58(3):587–593. https://doi.org/10.1016/j.ejpb.2004.04.002.TagedEn yethyl starch-coated polyplexes. Biomaterials. 2013;34(10):2530–2538.

TagedP 82. Secher-Hansen E, Langgard H, Schou J. Studies on the subcutaneous absorption in https://doi.org/10.1016/j.biomaterials.2012.12.025.TagedEn
mice. 3. Aseptic inflammation following subcutaneous injections. Acta Pharmacol TagedP108. Noga M, Edinger D, Ro € dl W, Wagner E, Winter G, Besheer A. Controlled shielding
Toxicol. 1967;25(3):299–306. https://doi.org/10.1111/j.1600-0773.1967.tb01436.x).TagedEn and deshielding of gene delivery polyplexes using hydroxyethyl starch (HES) and
TagedP 83. Yamaguchi K, Anderson JM. Biocompatibility studies of naltrexone sustained alpha-amylase. J Control Release. 2012;159(1):92–103. https://doi.org/10.1016/j.
release formulations. J Control Release. 1992;19(1-3):299–314. https://doi.org/ jconrel.2012.01.006.TagedEn
10.1016/0168-3659(92)90085-6.TagedEn TagedP109. Banerjee SS, Aher N, Patil R, Khandare J. Poly(ethylene glycol)-prodrug conju-
TagedP 84. Raghuvanshi A, Khan UA, Parveen U, Gupta A, Jain GK. An injectable sustained gates: concept, design, and applications. J Drug Deliv. 2012;2012:1–17. https://
release lipid based in situ gel system of aripiprazole for the management of doi.org/10.1155/2012/103973.TagedEn
schizophrenia. J Young Pharm. 2020;12(4):303–308. https://doi.org/10.5530/ TagedP110. Hrkach J, Langer R. From micro to nano: evolution and impact of drug delivery in
jyp.2020.12.82.TagedEn treating disease. Drug Deliv Transl Res. 2020;10(3):567–570. https://doi.org/
TagedP 85. Khan UA, Parveen U, Hasan N, et al. Parenteral sustained release lipid phase-tran- 10.1007/s13346-020-00769-6.TagedEn
sition system of ziprasidone: fabrication and evaluation for schizophrenia ther- TagedP111. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical
apy. Drug Des Dev Ther. 2020;14:2237–2247. https://doi.org/10.2147/dddt. applications. Adv Drug Deliv Rev. 2013;65(1):36–48. https://doi.org/10.1016/j.
S247196).TagedEn addr.2012.09.037.TagedEn
TagedP 86. Tam VH, Cohen DN, Ledesma KR, Guillory B, Chan K, Garey KW. Local tissue TagedP112. Burgess DJ, Crommelin DJA, Hussain AS, Chen M-L. Assuring quality and perfor-
response to subcutaneous administration of ceftriaxone in an animal model. Anti- mance of sustained and controlled release parenterals: EUFEPS workshop report.
microb Agents Chemother. 2020. https://doi.org/10.1128/AAC.02090-19.TagedEn AAPS PharmSci. 2004;6(1):E11. https://doi.org/10.1208/ps060111.TagedEn
TagedP 87. Schou J. Absorption of drugs from subcutaneous connective tissue. Pharmacol Rev. TagedP113. Iyer S, Barr W, Dance M, Coleman P, Karnes H. A ‘biorelevant’ system to investi-
1961;13(3):441–464.TagedEn gate in vitro drug released from a naltrexone implant. Int J Pharm. 2007;340(1-
TagedP 88. Van de Voorde J, Delaey C, Depypere H, Vanheel B. Mechanisms involved in the 2):104–118. https://doi.org/10.1016/j.ijpharm.2007.03.032.TagedEn
vasorelaxing influence of histamine on isolated human subcutaneous resistance TagedP114. Kinnunen HM, Sharma V, Contreras-Rojas LR, et al. A novel in vitro method to
arteries. Eur J Pharmacol. 1998;349(1):61–66. https://doi.org/10.1016/s0014- model the fate of subcutaneously administered biopharmaceuticals and associ-
2999(98)00179-4.TagedEn ated formulation components. J Control Release. 2015;214:94–102. https://doi.
TagedP 89. Benly P. Role of histamine in acute inflammation. J Pharma Sci Res. 2015;7 org/10.1016/j.jconrel.2015.07.016.TagedEn
(6):373–376.TagedEn TagedP115. Klose D, Azaroual N, Siepmann F, Vermeersch G, Siepmann J. Towards more
TagedP 90. Imakado S, Hamada K, Atsumi Y. Subcutaneous antihistamine injection is effec- realistic in vitro release measurement techniques for biodegradable micro-
tive to control a local allergic reaction to human insulin. Diabetes Res Clin Pract. particles. Pharm Res. 2009;26(3):691–699. https://doi.org/10.1007/s11095-
2009;85(3):e42–e43. https://doi.org/10.1016/j.diabres.2009.05.013.TagedEn 008-9747-4.TagedEn
TagedP 91. Farah S, Doloff JC, Mu € ller P, et al. Long-term implant fibrosis prevention in TagedP116. Bown HK, Bonn C, Yohe S, et al. In vitro model for predicting bioavailability of
rodents and non-human primates using crystallized drug formulations. Nat subcutaneously injected monoclonal antibodies. J Control Release. 2018;273:13–
Mater. 2019;18(8):892–904. https://doi.org/10.1038/s41563-019-0377-5.TagedEn 20. https://doi.org/10.1016/j.jconrel.2018.01.015.TagedEn
TagedP 92. Anderson JM, Niven H, Pelagalli J, Olanoff LS, Jones RD. The role of the fibrous cap- TagedP117. Jensen SS, Jensen H, Møller EH, et al. In vitro release studies of insulin from lipid
sule in the function of implanted drug-polymer sustained release systems. J implants in solution and in a hydrogel matrix mimicking the subcutis. Eur J Pharm
Biomed Mater Res. 1981;15(6):889–902. https://doi.org/10.1002/jbm.820150613.TagedEn Sci. 2016;81:103–112. https://doi.org/10.1016/j.ejps.2015.10.011.TagedEn
TagedP 93. Bartoli CR, Godleski JJ. Blood flow in the foreign-body capsules surrounding surgi- TagedP118. Shen J, Burgess DJ. In vitro−in vivo correlation for complex non-oral drug prod-
cally implanted subcutaneous devices. J Surg Res. 2010;158(1):147–154. https:// ucts: where do we stand? J Control Release. 2015;219:644–651. https://doi.org/
doi.org/10.1016/j.jss.2008.07.045.TagedEn 10.1016/j.jconrel.2015.09.052.TagedEn
TagedP 94. Vardar B, Kızılcı S. Incidence of lipohypertrophy in diabetic patients and a study TagedP119. Shen J, Burgess DJ. Accelerated in-vitro release testing methods for extended-
of influencing factors. Diabetes Res Clin Pract. 2007;77(2):231–236. https://doi. release parenteral dosage forms. J Pharm Pharmacol. 2012;64(7):986–996.
org/10.1016/j.diabres.2006.12.023.TagedEn https://doi.org/10.1111/j.2042-7158.2012.01482.x).TagedEn
TagedEnD. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508 1507

TagedP120. Jain RA, Rhodes CT, Railkar AM, Malick AW, Shah NH. Comparison of various TagedP143. Richter WF, Bhansali SG, Morris ME. Mechanistic determinants of biotherapeutics
injectable protein-loaded biodegradable poly(Lactide-co-glycolide) (PLGA) devi- absorption following SC administration. AAPS J. 2012;14(3):559–570. https://doi.
ces: in-situ-formed implant versus in-situ-formed microspheres versus isolated org/10.1208/s12248-012-9367-0.TagedEn
microspheres. Pharm Dev Technol. 2000;5(2):201–207. https://doi.org/10.1081/ TagedP144. Torres-Teran I, Ma rta V, Klein S. Prediction of subcutaneous drug absorption - do
pdt-100100535.TagedEn we have reliable data to design a simulated interstitial fluid? Int J Pharm.
TagedP121. Xie X, Lin W, Xing C, et al. In vitro and in vivo evaluations of PLGA microspheres 2021;610: 121257. https://doi.org/10.1016/j.ijpharm.2021.121257.TagedEn
containing nalmefene. PLoS One. 2015;10:(5) e0125953. https://doi.org/10.1371/ TagedP145. O’Farrell C, Stamatopoulos K, Simmons M, Hannah B. In vitro models to evaluate
journal.pone.0125953.TagedEn ingestible devices: Present status and current trends. Adv Drug Del Rev. 2021;178:
TagedP122. Kempe S, Metz H, Mader K. Do in situ forming PLG/NMP implants behave similar 113924. https://doi.org/10.1016/j.addr.2021.113924.TagedEn
in vitro and in vivo? A non-invasive and quantitative EPR investigation on the TagedP146. Kaewsichan L, Riyapan D, Prommajan P, Kaewsrichan J. Effects of sintering tem-
mechanisms of the implant formation process. J Control Release. 2008;130 peratures on micro-morphology, mechanical properties, and bioactivity of bone
(3):220–225. https://doi.org/10.1016/j.jconrel.2008.06.006.TagedEn scaffoldscontaining calcium silicate. ScienceAsia. 2011;37(3):240. https://doi.org/
TagedP123. Schliecker G, Schmidt C, Fuchs S, Ehinger A, Sandow J, Kissel T. In vitro and in vivo 10.2306/scienceasia1513-1874.2011.37.240.TagedEn
correlation of buserelin release from biodegradable implants using statistical TagedP147. Caracciolo G. Liposome−protein corona in a physiological environment:
moment analysis. J Control Release. 2004;94(1):25–37. https://doi.org/10.1016/j. Challenges and opportunities for targeted delivery of nanomedicines.
jconrel.2003.09.003.TagedEn Nanomed Nanotechnol Biol Med. 2015;11(3):543–557. https://doi.org/10.
TagedP124. Leung DH, Kapoor Y, Alleyne C, et al. Development of a convenient in vitro gel dif- 1016/j.nano.2014.11.003.TagedEn
fusion model for predicting the in vivo performance of subcutaneous parenteral TagedP148. ThermoFisher Scientific. Technical Resources: 24020 - HBSS, Calcium, Magnesium.
formulations of large and small molecules. AAPS PharmSciTech. 2017;18(6):2203– Waltham, MA: ThermoFisher Scientific; 2021.TagedEn
2213. https://doi.org/10.1208/s12249-016-0698-5.TagedEn TagedP149. Bou-Chacra N, Melo KJC, Morales IAC, et al. Evolution of choice of solubility and
TagedP125. Ma P, Li T, Xing H, et al. Local anesthetic effects of bupivacaine loaded lipid-poly- dissolution media after two decades of biopharmaceutical classification system.
mer hybrid nanoparticles: In vitro and in vivo evaluation. Biomed Pharmacother. AAPS J. 2017;19(4):989–1001. https://doi.org/10.1208/s12248-017-0085-5.TagedEn
2017;89:689–695. https://doi.org/10.1016/j.biopha.2017.01.175.TagedEn TagedP150. Do MP. In situ forming implants for the treatment of periodontitis Implants se for-
TagedP126. Zhong Z, Liu Z, Zhang X, et al. Effect of a controlled-release drug delivery system mant in situ pour le traitement des parodontites. Human health and pathology. Lille,
made of oleanolic acid formulated into multivesicular liposomes on hepatocellu- France: Universite  du Droit et de la Sante  - Lille II; 2014.TagedEn
lar carcinoma in vitro and in vivo. Int J Nanomed Vol. 2016;11:3111–3129. https:// TagedP151. Breitsamer M, Winter G. Needle-free injection of vesicular phospholipid gels-
doi.org/10.2147/ijn.s108445.TagedEn a novel approach to overcome an administration hurdle for semisolid depot
TagedP127. Park TG, Lu W, Crotts G. Importance of in vitro experimental conditions on pro- systems. J Pharm Sci. 2017;106(4):968–972. https://doi.org/10.1016/j.xphs.
tein release kinetics, stability and polymer degradation in protein encapsulated 2016.12.020.TagedEn
poly (d,l-lactic acid-co-glycolic acid) microspheres. J Control Release. 1995;33 TagedP152. Usach I, Martinez R, Festini T, Peris J-E. Subcutaneous injection of drugs: literature
(2):211–222. https://doi.org/10.1016/0168-3659(94)00084-8.TagedEn review of factors influencing pain sensation at the injection site. Adv Ther.
TagedP128. Otte A, Damen F, Goergen C, Park K. Coupling the in vivo performance to the in 2019;36(11):2986–2996. https://doi.org/10.1007/s12325-019-01101-6.TagedEn
vitro characterization of PLGA microparticles. Int J Pharm. 2021;604: 120738. TagedP153. Semmling B, Nagel S, Sternberg K, Weitschies W, Seidlitz A. Long-term stable
https://doi.org/10.1016/j.ijpharm.2021.120738.TagedEn hydrogels for biorelevant dissolution testing of drug-eluting stents. J Pharma
gedP1 Ta 29. Heo JY, Noh JH, Park SH, et al. An injectable click-crosslinked hydrogel that Technol Drug Res. 2013;2(1). https://doi.org/10.7243/2050-120X-2-19.TagedEn
prolongs dexamethasone release from dexamethasone-loaded microspheres. TagedP154. Webb P. Temperatures of skin, subcutaneous tissue, muscle and core in resting
Pharmaceutics. 2019;11(9):438. https://doi.org/10.3390/pharmaceu- men in cold, comfortable and hot conditions. Eur J Appl Physiol Occup Physiol.
tics11090438.TagedEn 1992;64(5):471–476. https://doi.org/10.1007/bf00625070.TagedEn
TagedP130. Liu Q, Zhang H, Zhou G, et al. In vitro and in vivo study of thymosin alpha1 biode- TagedP155. Dokoumetzidis A, Macheras P. A century of dissolution research: From Noyes and
gradable in situ forming poly(lactide-co-glycolide) implants. Int J Pharm. Whitney to the biopharmaceutics classification system. Int J Pharm. 2006;321(1-
2010;397(1-2):122–129. https://doi.org/10.1016/j.ijpharm.2010.07.015.TagedEn 2):1–11. https://doi.org/10.1016/j.ijpharm.2006.07.011.TagedEn
TagedP131. Eliaz RE, Wallach D, Kost J. Delivery of soluble tumor necrosis factor receptor from TagedP156. Rawat A, Bhardwaj U, Burgess DJ. Comparison of in vitro−in vivo release of
in-situ forming PLGA implants: in-vivo. Pharm Res. 2000;17(12):1546–1550. RisperdalÒ ConstaÒ microspheres. Int J Pharm. 2012;434(1-2):115–121. https://
https://doi.org/10.1023/a:1007621512647.TagedEn doi.org/10.1016/j.ijpharm.2012.05.006.TagedEn
TagedP132. Iyer S, Barr W, Karnes H. A ‘biorelevant’ approach to accelerated in vitro drug TagedP157. Kozak J, Rabiskova M, Lamprecht A. In-vitro drug release testing of parenteral for-
release testing of a biodegradable, naltrexone implant. Int J Pharm. 2007;340(1- mulations via an agarose gel envelope to closer mimic tissue firmness. Int J Pharm.
2):119–125. https://doi.org/10.1016/j.ijpharm.2007.03.033.TagedEn 2021;594: 120142. https://doi.org/10.1016/j.ijpharm.2020.120142.TagedEn
TagedP133. Iyer SS, Barr WH, Karnes HT. Characterization of a potential medium for ‘biorele- TagedP158. Schreiner V, Detampel P, Jirkof P, Puchkov M, Huwyler J. Buprenorphine
vant’ in vitro release testing of a naltrexone implant, employing a validated sta- loaded PLGA microparticles: characterization of a sustained-release formula-
bility-indicating HPLC method. J Pharm Biomed Anal. 2007;43(3):845–853. tion. J Drug Delivery Sci Technol. 2021;63: 102558. https://doi.org/10.1016/j.
https://doi.org/10.1016/j.jpba.2006.08.023.TagedEn jddst.2021.102558.TagedEn
TagedP134. Kortesuo P, Ahola M, Kangas M, Kangasniemi I, Yli-Urpo A, Kiesvaara J. In vitro TagedP159. Xie L, Beyer S, Vogel V, Wacker MG, M€antele W. Assessing the drug release from
evaluation of sol−gel processed spray dried silica gel microspheres as carrier in nanoparticles: overcoming the shortcomings of dialysis by using novel optical
controlled drug delivery. Int J Pharm. 2000;200(2):223–229. (https://doi.org/ techniques and a mathematical model. Int J Pharm. 2015;488(1):108–119.
10.1016/s0378-5173(00)00393-8).TagedEn https://doi.org/10.1016/j.ijpharm.2015.03.080.TagedEn
TagedP135. Modh H, Fang DJ, Ou YH, et al. Injectable drug delivery systems of doxorubicin TagedP160. Wacker MG, Lu X, Burke M, Nir I, Fahmy R, Zaidi K 2021 (Accepted for public con-
revisited: in vitro-in vivo relationships using human clinical data. manuscript sultation). Testing the in-vitro product performance of nanomaterial-related
submitted Int J Pharm. 2021.TagedEn drug products: View of the USP Expert Panel Pharmacop Forum 47(6).TagedEn
TagedP136. He Q, Shi J, Zhu M, Chen Y, Chen F. The three-stage in vitro degradation behavior TagedP161. Thati S, McCallum M, Xu Y, et al. Novel applications of an in vitro injection model
of mesoporous silica in simulated body fluid. Microporous Mesoporous Mater. system to study bioperformance: case studies with different drug modalities. J
2010;131(1-3):314–320. https://doi.org/10.1016/j.micromeso.2010.01.009.TagedEn Pharm Innov. 2020;15(2):268–280. https://doi.org/10.1007/s12247-020-09437-1.TagedEn
TagedP137. Choi Y, Lee JE, Lee JH, Jeong JH, Kim J. A biodegradation study of SBA-15 micropar- TagedP162. Mertz N, Østergaard J, Yaghmur A, Larsen SW. Transport characteristics in a
ticles in simulated body fluid and in vivo. Langmuir. 2015;31(23):6457–6462. novel in vitro release model for testing the performance of intra-articular
https://doi.org/10.1021/acs.langmuir.5b01316.TagedEn injectables. Int J Pharm. 2019;566:445–453. https://doi.org/10.1016/j.
TagedP138. Marques MRC, Loebenberg R, Almukainzi M. Simulated biological fluids with pos- ijpharm.2019.04.083.TagedEn
sible application in dissolution testing. Dissol Technol. 2011;18(3):15–28. https:// TagedP163. Nothnagel L, Wacker MG. How to measure release from nanosized carriers? Eur J
doi.org/10.14227/dt180311p15.TagedEn Pharm Sci. 2018;120:199–211. https://doi.org/10.1016/j.ejps.2018.05.004.TagedEn
TagedP139. Barbeck M, Serra T, Booms P, et al. Analysis of the in vitro degradation and the in TagedP164. Janas C, Mast M-P, Kirsamer L, et al. The dispersion releaser technology is an
vivo tissue response to bi-layered 3D-printed scaffolds combining PLA and effective method for testing drug release from nanosized drug carriers. Eur J
biphasic PLA/bioglass components − Guidance of the inflammatory response as Pharm Biopharm. 2017;115:73–83. https://doi.org/10.1016/j.ejpb.2017.02.006.TagedEn
basis for osteochondral regeneration. Bioact Mater. 2017;2(4):208–223. https:// TagedP165. Zolnik BS, Raton J-L, Burgess DJ. Application of USP apparatus 4 and in situ fiber
doi.org/10.1016/j.bioactmat.2017.06.001.TagedEn optic analysis to microsphere release testing. Dissol Technol. 2005;12(2):11–14.
TagedP140. Uemura A, Ogawa S, Isono Y, Tanaka R. Elucidation of the time-dependent degra- https://doi.org/10.14227/dt120205p11.TagedEn
dation process in insoluble hyaluronic acid formulations with a controlled degra- TagedP166. Nicolaides E, Symillides M, Dressman JB, Reppas C. Biorelevant dissolution testing
dation rate. J Tissue Eng. 2019;10: 204173141988503. https://doi.org/10.1177/ to predict the plasma profile of lipophilic drugs after oral administration. Pharm
2041731419885032.TagedEn Res. 2001;18(3):380–388. https://doi.org/10.1023/a:1011071401306.TagedEn
TagedP141. Kokubo T, Takadama H. How useful is SBF in predicting in vivo bone bioac- TagedP167. Singh I, Aboul-Enein HY. Advantages of USP Apparatus IV (Flow-through Cell
tivity? Biomaterials. 2006;27(15):2907–2915. https://doi.org/10.1016/j.bioma- Apparatus) in dissolution studies. J Iran Chem Soc. 2006;3:220–222. https://doi.
terials.2006.01.017.TagedEn org/10.1007/BF03247211.TagedEn
TagedP142. Jamuna-Thevi K, Suleiman MJ, Sabri SN. The in vitro degradation of PLGA/nanoa- TagedP168. Gietz U, Arvinte T, Mader E, Oroszlan P, Merkle HP. Sustained release of injectable
patite/lauric acid composite membrane: a comparative study in phosphate buffer zinc-recombinant hirudin suspensions: development and validation of in vitro
saline and simulated body fluid. Macromol Symp. 2017;371(1):101–106. https:// release model. Eur J Pharm Biopharm. 1998;45:259–264. https://doi.org/10.1016/
doi.org/10.1002/masy.201600048.TagedEn S0939-6411(98)00008-3.TagedEn
TagedEn1508 D. Li et al. / Journal of Pharmaceutical Sciences 112 (2023) 1492−1508

TagedP169. Solorio L, Babin BM, Patel RB, Mach J, Azar N, Exner AA. Noninvasive characteriza- TagedP175. Larsen SW, Østergaard J, Yaghmur A, Jensen H, Larsen C. Use of in vitro release
tion of in situ forming implants using diagnostic ultrasound. J Control Release. models in the design of sustained and localized drug delivery systems for subcu-
2010;143(2):183–190. https://doi.org/10.1016/j.jconrel.2010.01.001.TagedEn taneous and intra-articular administration. J Drug Deliv Sci Technol. 2013;23
TagedP170. Li Z, Mu H, Larsen SW, Jensen H, Østergaard J. Initial leuprolide acetate (4):315–324. https://doi.org/10.1016/S1773-2247(13)50048-7.TagedEn
release from poly(d,l-lactide-co-glycolide) in situ forming implants as stud- TagedP176. Alam J, Mcallister A, Scaramucci J, Jones W, Rogge M. Pharmacokinetics and
ied by ultraviolet-visible imaging. Mol Pharm. 2020;17(12):4522–4532. pharmacodynamics of interferon Beta-1a (IFN??-1a) in healthy volunteers
https://doi.org/10.1021/acs.molpharmaceut.0c00625.TagedEn after intravenous, subcutaneous or intramuscular administration. Clin Drug
TagedP171. Østergaard J. UV imaging in pharmaceutical analysis. J Pharm Biomed Anal. Investig. 1997;14(1):35–43. https://doi.org/10.2165/00044011-199714010-
2018;147:140–148. https://doi.org/10.1016/j.jpba.2017.07.055.TagedEn 00005.TagedEn
TagedP172. Ye F, Larsen SW, Yaghmur A, Jensen H, Larsen C, Ostergaard J. Drug release into TagedP177. Sierra-Ramírez JA, Lara-Ricalde R, Lujan M, et al. Comparative pharmacoki-
hydrogel-based subcutaneous surrogates studied by UV imaging. J Pharm Biomed netics and pharmacodynamics after subcutaneous and intramuscular admin-
Anal. 2012;71:27–34. https://doi.org/10.1016/j.jpba.2012.07.024.TagedEn istration of medroxyprogesterone acetate (25 mg) and estradiol cypionate (5
TagedP173. Kempe S, Metz H, Pereira PG, Ma €der K. Non-invasive in vivo evaluation of in situ mg). Contraception. 2011;84(6):565–570. https://doi.org/10.1016/j.contracep-
forming PLGA implants by benchtop magnetic resonance imaging (BT-MRI) and tion.2011.03.014.TagedEn
EPR spectroscopy. Eur J Pharm Biopharm. 2010;74(1):102–108. https://doi.org/ TagedP178. Kuboyama K, Tanga N, Suzuki R, Fujikawa A, Noda M. Protamine neutralizes
10.1016/j.ejpb.2009.06.008.TagedEn chondroitin sulfate proteoglycan-mediated inhibition of oligodendrocyte differ-
TagedP174. Zheng F, Xiong W, Sun S, Zhang P, Zhu JJ. Recent advances in drug release monitoring. entiation. PLoS One. 2017;12:(12): e0189164. https://doi.org/10.1371/journal.
Nanophotonics. 2019;8(3):391–413. https://doi.org/10.1515/nanoph-2018-0219.TagedEn pone.0189164.TagedEn

You might also like