Submit manuscript...
Journal of
eISSN: 2473-0831

Analytical & Pharmaceutical Research

Review Article Volume 2 Issue 5

Kidney-Specific Drug Delivery: Review of Opportunities, Achievements, and Challenges

Dikran Sarko, Rania B Georges

Correspondence: Dikran Sarko, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Baath University-P.O.Box 77, Homs, Syria, Tel +963 31 9910

Received: June 25, 2016 | Published: July 5, 2016

Citation: Sarko D, Georges RB (2016) Kidney-Specific Drug Delivery: Review of Opportunities, Achievements, and Challenges. J Anal Pharm Res 2(5): 00033. DOI: 10.15406/japlr.2016.02.00033

Download PDF

Abstract

Kidneys are involved in essential complex procedures that maintain the body in equilibrium and preservethe interior environment necessary for life. Nephritis and nephrosis were ranked among the leading causes of death in all ages in the last global burden of disease assessment of the World Health Organization (WHO). Regarding kidney disease therapeutics, the unfavourable extra-target effects, the narrow therapeutic index, the inactivation of the drug before reaching its target, or the affected normal distribution due to the pathophysiology of diseases, all of these conducted to the requirement of new therapeutic aspects. An efficient kidney-specific drug delivery system will serve as fascinating approach and attractive option conquering such problems, improving the therapeutic index, and direct to a favourable pharmacokinetic profile of drugs. Such breakthrough in renal targeting will provide an efficacious major key for controlling the incidence of those problems, affording a logical treatment methodology managing all at-risk patients, representing a major intention for therapy and impart an attractive pharmacological implement to elucidate the mechanisms of drug action in the kidney.Several attempts to achieve the optimal renal delivery systems had been investigated; low-molecular-weight-proteins (LMWPs), low-molecular-weight chitosan (LMWC), Poly(vinylpyrrolidone-co-dimethyl maleic acid) (PVD), anionized polyvinylpyrrolidone (PVP), galectin-3 carbohydrate recognition domain (G3-C12), and most recently epsilon poly-l-lysine-derivatives (εPLL) and the carrier peptide (KKEEE)3K. The majority of kidney-specific delivery systems target the proximal tubular cells. The specific uptake of the carriers by renal proximal tubular cells is attributed in many cases to megalin-mediated endocytosis. Moreover, the overall charge of the carrier seems to play a key role in kidney-specific drug delivery. On the other hand, mesangial cells represent a particularly suitable target for drug delivery by particulate drug delivery systems such as nanoparticles or liposomes taking into account the particle diameter. In this review, we summarize briefly the aforementioned issues.

Keywords: Renal targeting; Renal proximal tubule cells; Megalin; Mesangium

Abbreviations

WHO: World Health Organization; KD: Kidney Diseases; DDS: Drug Delivery System; LMWP: Low-Molecular-Weight-Proteins; ADEPT: Antibody-Directed Enzyme Prodrug Therapy; PEG: Polyethylene Glycol; PLL: Poly-L-Lysine; LDL: Low-Density Lipoproteins; HDL: High-Density Lipoproteins; PLGA: Poly(d,l-Lactic-Coglycolic-Acid); FDA: Food and Drug Administration; CKD: Chronic Kidney Diseases; ADPKD: Autosomal Dominant Polycystic Kidney Disease; LMWC: Low Molecular Weight Chitosan; PVD: Poly(vinylpyrrolidone-co-dimethyl maleic acid);PVP: polyvinylpyrrolidone;DOTA: 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid; εPLL: Epsilon Poly-l-Lysine; OX7-IL: liposomes with Fab′ fragments of OX7 mAb

Introduction

Drug targeting

Drug-targeting aims at reducing the obstacles caused by extra-target effects, the narrow therapeutic index and the inactivation of the drug before reaching its target. Unfortunately, most of the pharmaceutics used today lack a selective delivery into their target tissues. The selective delivery or selective activation in the targeted tissue minimizes toxic side effects [1]. The improvement of the pharmacokinetic profile should be accomplished by drug targeting strategies [2,3]. However, diverse drug-targeting concepts have been pinpointed in order to fulfill those principles [4,5].

Prodrugs are derivatives of drug molecules that undergo an enzymatic or chemical transformation in vivo to release the active parent drug, which can then exert the desired pharmacological effect [6]. Therefore, the main idea here is the selective liberation of the pharmacological active part of the prodrug in the target tissue depending on tissue-specific metabolic pathways such as tissue-specific enzyme-depending release [7], altered physiological characteristics such as pH sensitive conjugates [8], agents activated by hypoxia [9], or antibody-directed enzyme prodrug therapy (ADEPT) [10]. In drug delivery, the achievement of tissue-selective or site-selective drug delivery systems (DDSs) is of great interest particularly in the prodrug-based approach [6,11].

Drug delivery systems

A variety of carriers such as viral vectors [12], colloidal particles or macromolecular carriers (such as liposomes [13], nanoparticles [14], microspheres [15], lipid particles and polymeric micelles [16-18]), modified-plasma proteins, polysaccharides [19], biodegradable carriers [20], dendrimers [21], antibodies [22], and peptide carriers have been developed [23].

A large diversity of drug delivery systems has been developed to enhance the therapeutic effect in the target tissue. The exclusive transfer of a drug to the targeted site of action that has been accomplished with minimal toxic side effects, and the usage of a pharmacologically inactive vector are main features of an ideal carrier for drug-targeting delivery system [24]. A specifically targeting drug delivery system can serve not only as a therapeutic vehicle but also as a research tool. Generally, the drug-targeting approach depends on the drug to be delivered, the target tissue, and of course the proper delivery system.

Carriers could be classified into three major types; the particle-type, the soluble and the cellular carriers. Liposomes, nanoparticles, microspheres, lipid particles, and polymeric micelles are all included in the particle-type carriers. Whereas peptides, modified-plasma proteins, polysaccharides, and biodegradable carriers are classified under soluble carriers. However, viral vectors belong to the cellular carriers. Despite the advantages provided by the cellular carriers depending upon their natural biocompatibility and their possibility to cause an immunological response is still a hurdle.

Polymers could be classified according to their natural or synthetic origin, stability (whether biodegradable or not), backbone, and upon their chemical nature (vinyl and acrylic polymers, polyethylene glycol (PEG), polysaccharides, polyamino acids, etc).[8] The natural polymer carriers include several polysaccharides (dextrans, inulin, or chitosan), proteins (albumin) or glycoproteins (transferrin), as well as cationic polymeric carriers such as PLL (poly-l-lysine) backbones [25].

In addition to the size of liposomes (ranging from 20 to 10,000 nm), factors such as charge and lipid composition can extensively dominate their behavior in vivo. Liposomes are exploited for macrophage-specific delivery thus involved in passive drug targeting which permits the sustained release of drug over time. Usually, modifications of the surface by a targeting device, homing ligand, or by PEGylation improve their in vivo behaviors [26,27]. Size-expansion strategies limit their extravasation, thereby minimizes the drug distribution to non-target sites. PEGylation is involved in the prolonged circulation time of liposomes by preventing their recognition by phagocytes and increasing their peripheral distribution [28].

Similar principles could be achieved depending on the endogenous lipid particles such as low- or high-density lipoproteins (LDL/HDL) which can serve as “natural targeted liposomes” [29]. New applications and opportunities for intracellular delivery of various molecules such as small interfering ribonucleic acid (siRNA), antisense oligonucleotides (ASOs), recombinant proteins and cloned genes, have been achieved through advances in liposome design. In addition, several studies have been performed on the delivery of anti-fungal, anti-viral and anti-cancer drugs by liposome formulations in humans; such as amphotericin B, acyclovir and doxorubicin, respectively. On the other hand, several challenges and obstacles are yet to be faced and defeated; high production costs, short half-life, low solubility, and the possibility of encapsulated drug’s leakage and fusion [30].

Nanoparticles and microspheres can be assigned either to the soluble or to the particle type carriers. The main backbone of these carriers is based on diversity of polymers such as dextran, ficoll, sepharose, or PLL. Nowadays, poly(D,L-lactic-co-glycolic-acid) (PLGA) microspheres have been widely studied to gain a wide acceptance for application as nanoparticles and microspheres following to their approval for use in humans by the American Food and Drug Administration (FDA) [31]. Parenteral application of microspheres and nanoparticles for the cell selective delivery of drugs is not the only administration route since they have been studied more recently for their application in oral and pulmonary delivery of peptides and peptidomimetics [20]. Antibodies are aimed at targeting tumor-associated antigens that are over-expressed by tumor cells. Therefore, the birth of new techniques such as recombinant DNA and protein engineering has led to the development of optimal tailored-antibodies [22]. The stretch of amino acids or peptides, contained by a biomolecule which is responsible for specific receptor binding, is called the homing-ligand. Their covalent attachment to a carrier backbone can result in targeted DDS [32]. Polymeric micelles are small (10-100nm) in size and they have a core-shell structure. In contrast to their hydrophobic core, the shell represents their hydrophilic part. Micelles provide a penetration property inside the tissue for targeted DDS. Generally, their utility is still challenging since they disintegrate rapidly in vivo [33]. Usually, the application of peptide carriers requires an over-expressed receptor and sufficient in vivo stability. Additionally, the conjugated drug should not interfere with the binding region of the peptide. Peptides were developed in different ways, for example for targeted transport using carrier peptides for tumor and tissue targeting [34,35].

The systemic side effects of therapeutic agents on healthy and non-targeted tissues are one of the most serious problems particularly in oncology and gene therapy. Many of the existing pharmaceutics have their limitations due to the lack of selective delivery into their target tissues. Consequently, site/organ-specific drug targeting is an attractive strategy to reduce unwanted side effects and to enhance drug efficacy within the targeted tissue [36,37]. Here we focus on kidney targeting with a collection of reviews and perspectives that highlights some of the main strategies in this field.

Discussion

Kidney-specific drug delivery

The major functions of the kidney are the maintenance of body water and sodium balance, the filtration of waste products from the bloodstream, the secretion of hormones, and multiple homeostatic controls such as the acid-base regulation of the blood. The current number of 280 million patients suffering from chronic kidney diseases (CKD) increases incessantly. The costs for the treatment of kidney diseases are predicted to exceed 1 trillion US$ over the next decade. Drugs for treating kidney diseases are often limited by tolerability and extra-kidney safety concerns, which prevent their use at maximally effective doses. Targeting drugs to the kidneys may circumvent these limitations and reduce toxicity of new, established, and pre-existing drugs. It has therefore evolved as a cherished but elusive goal in pharmaceutical sciences. For examples, long-term therapy of CKD is accompanied by serious side effects and numerous promising drug candidates failed during clinical trials due to safety issues and lack of efficacy, e.g., bardoxolone methyl and paricalcitol in CKD, avosentan in diabetic nephropathy,and sirolimus and everolimus in autosomal dominant polycystic kidney disease (ADPKD) [38]. Covalently linked drug-polymers can be applied to improve the therapeutic index of toxic drugs. To prevent the shift in cytotoxicity pattern resulting from the delivery system, biodegradable delivery systems such as peptide-based carriers were preferred [39]. Another approach makes use of some endogenous enzymes having relatively high concentrations in kidneys such as amino acid l-decarboxylase and γ-glutamyl transpeptidase [40]. Based on this, substrates of these enzymes to chemically modify drugs had been prepared and used in the hope that drug would be released in proximal tubular cells via the relevant enzyme.

Consequently, efforts have been made to enable a specific uptake of drugs in the kidneys [40]. Attempts to use small protein carriers such as lysozymes were hampered by their renal toxicity and cardiovascular side effects [41]. Prodrugs that are activated by kidney-associated enzymes showed low accumulation rates in the kidneys [42]. Streptavidin conjugates that target the high levels of endogenous biotin in the kidneys failed due to unfavourable pharmacokinetics of this high molecular weight protein [43]. Liposomal formulations [44], dendrimer conjugates [21] as well as copolymer conjugates [45] also did not yet exert the properties required. Relatively few papers on drug delivery or targeting research however appear in the top ranking scientific journals, probably because the concepts or technologies presented are often not recognized as most innovative scientifically but rather application oriented [46].

The majority of kidney-specific delivery systems target the proximal tubular cells and may contribute in the treatment of renal diseases such as kidney transplantation, ureteral obstruction, diabetes, proteinuria, and in some diseases involving changes in renal tubular function such as Fanconi and Bartter´s syndrome.

Alkylglycoside and sugar-modified low-molecular-weight peptides: Alkylglycoside and sugar-modified low-molecular-weight peptides have been shown to have renal targeting potential in vivo. In 1999, Suzuki et al. [47] had suggested a novel transport mechanism in the kidneys that can be used for the specific renal delivery of glycosylated peptides [47]. Their observations reveal the saturation of the renal uptake in vivo with increasing dose, and there is an effective mechanism for uptake from blood. Moreover, they had proposed that the renal uptake may not be dependent on derivatives having a cationic nature. However, Shirota et al. [48] had suggested that the anionic moiety could reduce the renal targeting efficiency [48]. Thus, the targeting efficacy of the alkylglucoside vector seems to depend on, at least, the size and charge of the ligand that it delivers.

Low molecular weight proteins (LMWPs)

LMWPs involve active proteins in the circulatory system such as enzymes (lysozymes), immune proteins (such as light chain immunoglobulins) and peptide hormones (such as insulin). However, lysozyme represents the most widely studied LMWP. The most extensively studied kidney-targeted lysozyme-conjugates are with naproxen (peptide bond) [49-51], triptolide (ester linkage) [52,53], captopril (disulfide bond) [54-57], and anticancer agents [1,58]. Moreover, a number of other drugs have been linked to lysozyme or various carriers in several ways [59,60].

LMWPs can be filtered at the glomerulus and reabsorbed in the renal tubules. Generally, the kinetics of the macromolecular carrier over rule the intrinsic kinetics of the drug. Therefore, a drug-macromolecular carrier conjugate is rapidly cleared from the blood supply and undergoes drug release and activation in lysosomes. Normally, distribution of the released drug in the kidney is relatively slow allowing more concentration of drug in the kidneys relative to plasma [61-64]. However, due to the diversity and variety of the functional groups presented on the LMWP, conjugation procedures should be performed carefully to avoid the very common self-aggregation and degeneration. Additionally, attempts to use LMWPs such as lysozymes were hampered by their renal toxicity and cardiovascular side effects [41].

Low molecular weight chitosan (LMWC)

Chitosan has been widely used in drug delivery systems due to its excellent biocompatibility and biodegradability. It is derived from chitin and consists of glucosamine and N-acetyl-glucosamine. However, different degrees of acetylation and molecular weights of LMWC had been investigated for their kidney-specific drug delivery [65,66]. LMWC is specifically taken up by renal tubular cells probably via megalin [67-69].  However, LMWC was cleared from the kidneys more rapidly in comparison with lysozyme, therefore, it has safer pharmacokinetic profile.

Chitosan oligomers can be a potential drug carrier for renal targeting delivery. It was used as a carrier for zidovudine (AZT), since the later has a very short half-life and is eliminated very quickly in human plasma and kidney after administration [70]. However, more characterization and investigation worth to be performed.

Anionized polyvinylpyrrolidone (PVP) and Poly(vinylpyrrolidone-co-dimethyl maleic acid) (PVD): PVD had been reported as a carrier for kidney-specific drug delivery. However, the polymer needs long time (24h) after i.v. injection in order to accumulate only 80% specifically in the kidneys. Further study in mice demonstrates that there is a relationship between the molecular weight and the charge of PVD derivatives regarding their renal accumulations. Additionally, carboxylated PVP was taken up by the renal proximal tubular epithelial cells in vivo after i.v. injection [71,72].

G3-C12 peptide: The galectin-3 carbohydrate recognition domain (G3-C12) was shown to specifically accumulate in mouse kidneys after i.v. injection [73]. The peptide-captopril conjugate was evaluated with a disulfide bond which can be cleaved by reduced glutathione in the kidney. The peptide–drug conjugate accumulated specifically in the kidney soon after i.v. injection into mice, and the accumulation had been attributed largely to the reabsorption of the peptide by the renal proximal tubule cells [74].

Epsilon Poly-l-lysine-Derivatives (εPLL) and The Carrier Peptide (KKEEE)3K: Recently, Mier and co-workers had started a development program for a kidney-specific carrier peptide. [75] Based on the effect of lysine which interacts with receptors on the apical side of proximal tubule cells [76,77], DOTA-εPLL showed very exclusive accumulation in the kidneys. Due to the structure of DOTA-εPLL comprising unnatural peptide bonds, DOTA as an additional compound, and a long retention in the kidneys, a peptide consisting only of natural amino acids and standard peptide linkages was developed. Based on their findings, the authors stated that a balanced ratio between positively and negatively charged functional groups is obviously essential to achieve high kidney specificity. Different peptides were synthesized to identify a carrier with improved clearance properties and balanced ratio between positive and negative charged functional groups. The ideal peptide not containing any unnatural residues was found to be (KKEEE)3K [75]. The results obtained demonstrate an indication of megalin-mediated endocytosis of (KKEEE)3K into proximal tubule cells. The carrier peptide (KKEEE)3K can circumvent the specific disadvantages of LMWP and other macromolecular carriers. It shows high kidney selectivity, even with conjugated drugs (e.g. ciprofloxacin), rapid kidney accumulation, renal clearance within a few hours, and has a nontoxic profile. Therefore, (KKEEE)3K is a promising carrier for renal targeting.

Particulate kidney-targeted drug delivery systems: Usually, particulate drug delivery systems such as nanoparticles or liposomes have been omitted in renal targeting due to their large size and the limitation of glomerular filtration. They are being investigated for numerous medical applications and are showing potential as an emerging class of carriers for drug delivery. Despite proven success in their accumulation at a selected few organs such as tumor and liver, reports on their effective delivery to other organs still remain scarce. Choi et al. [78] showed that PEGylated gold nanoparticlesof ~75 ± 25-nm diameters target the mesangium of the kidney demonstrating the effects of particle diameter on targeting the mesangium [78]. This finding establishes design criteria for constructing nanoparticle-based delivery systems for targeting diseases that involve the mesangium. Moreover, this finding is of special interest since the majority of kidney-specific drug delivery systems are targeting the proximal tubular cells rather than the mesangium. On the other hand, Tuffin et al. [79] had targeted the mesangium by preparing liposomes with Fab′ fragments of OX7 mAb (OX7-IL) [79]. However, rats that were given a single intravenous injection of low-dose doxorubicin encapsulated in OX7-IL showed extensive glomerular damage.

Conclusion

Kidney diseases require, usually, long-term administration of therapeutics; therefore, they are always accompanied with systemic toxicities and adverse side effects. Consequently, it is necessary to develop a kidney-targeted drug delivery system in order to overcome these obstacles .Several attempts have been performed aiming to target the kidneys, and the majority of kidney-specific delivery systems targets the proximal tubular cells and may contribute to the treatment of renal diseases. Such strategies involve LMWPs, PVP, PVD, LMWC, G3-C12 peptide, εPLL derivatives and the carrier peptide (KKEEE)3K. The specific uptake by renal proximal tubular cells is confirmed in many cases to bemegalin-mediated endocytosis. Additionally, the overall charge of the carrier seems to play a key role in kidney-specific drug delivery. On the other hand, particulate drug delivery systems such as nanoparticles or liposomes seem to be useful in targeting diseases that involve the mesangium. However, the particle diameter is crucial for targeting the mesangium. Finally, the challenge remains in translating the use of those carriers in clinical trials and applications.

References

  1. Ding S, Bierbach U (2015) Target-selective delivery and activation of platinum-based anticancer agents. Future Med Chem 7(7): 911-927.
  2. Mickan A, Sarko D, Haberkorn U, Mier W (2014) Rational design of CPP-based drug delivery systems: considerations from pharmacokinetics. Curr Pharm Biotechnol 15(3): 200-209.
  3. Sarko D, Beijer B, Garcia RB, Nothelfer EM, Leotta K, et al. (2010) The pharmacokinetics of cell-penetrating peptides. Mol Pharm 7(6): 2224-2231.
  4. Mier W, Hoffend J, Haberkorn U, Eisenhut M (2005) Current Strategies in Tumor-Targeting. In: Marek Los & Spencer B Gibson (Eds.), Springer US, New York, USA, pp. 343-355.
  5. Melisi D, Rimoli MG (2011) Recent developments in prodrug design: drug targeting, pharmacological and pharmacokinetic improvements related to a reduction of adverse effects. Curr Top Med Chem 11(18): 2264.
  6. Rautio J, Kumpulainen H, Heimbach T, Oliyai R, Oh D, et al. (2008) Prodrugs: design and clinical applications. Nat Rev Drug Discov 7(3): 255-270.
  7. Liederer BM, Borchardt RT (2006) Enzymes involved in the bioconversion of ester-based prodrugs. J Pharm Sci 95(6): 1177-1195.
  8. Ulbrich K, Subr V (2004) Polymeric anticancer drugs with pH-controlled activation. Adv Drug Deliv Rev 56(7): 1023-1050.
  9. de Groot FM, Damen EW, Scheeren HW (2001) Anticancer prodrugs for application in monotherapy: targeting hypoxia, tumor-associated enzymes, and receptors. Curr Med Chem 8(9): 1093-1122.
  10. Bagshawe KD, Sharma SK, Begent RH (2004) Antibody-directed enzyme prodrug therapy (ADEPT) for cancer. Expert Opin Biol Ther 4(11): 1777-1789.
  11. Ettmayer P, Amidon GL, Clement B, Testa B (2004) Lessons learned from marketed and investigational prodrugs. J Med Chem 47(10): 2393-2404.
  12. Ewer KJ, Lambe T, Rollier CS, Spencer AJ, Hill AV, et al. (2016) Viral vectors as vaccine platforms: from immunogenicity to impact. Curr Opin Immunol 41: 47-54.
  13. Nguyen TX, Huang L, Gauthier M, Yang G, Wang Q (2016) Recent advances in liposome surface modification for oral drug delivery. Nanomedicine 11(9): 1169-1185.
  14. Majzoub RN, Ewert KK, Safinya CR (2016) Cationic liposome-nucleic acid nanoparticle assemblies with applications in gene delivery and gene silencing. Philos Trans A Math Phys Eng Sci 374(2072): 20150129.
  15. Andhariya JV, Burgess DJ (2016) Recent advances in testing of microsphere drug delivery systems. Expert Opin Drug Deliv 13(4): 593-608.
  16. Tanbour R, Martins AM, Pitt WG, Husseini GA (2016) Drug Delivery Systems Based on Polymeric Micelles and Ultrasound: A Review. Curr Pharm Des 22(19): 2796-2807.
  17. Gothwal A, Khan I, Gupta U (2016) Polymeric Micelles: Recent Advancements in the Delivery of Anticancer Drugs. Pharm Res 33(1): 18-39.
  18. Biswas S, Kumari P, Lakhani PM, Ghosh B (2016) Recent advances in polymeric micelles for anti-cancer drug delivery. Eur J Pharm Sci 83: 184-202.
  19. Maruyama A, Ishihara T, Kim JS, Kim SW, Akaike T (1997) Nanoparticle DNA Carrier with Poly(l-lysine) Grafted Polysaccharide Copolymer and Poly(d,l-lactic acid). Bioconjugate Chem 8(5): 735-742.
  20. Farahidah M, van der WCF (2008) Engineering biodegradable polyester particles with specific drug targeting and drug release properties. J Pharm. Sci 97(1): 71-87.
  21. Kaminskas LM, Kelly BD, McLeod VM, Boyd BJ, Krippner GY, et al. (2009) Pharmacokinetics and Tumor Disposition of PEGylated, Methotrexate Conjugated Poly-l-lysine Dendrimers. Mol Pharm 6(4): 1190-1204.
  22. Schaedel O, Reiter Y (2006) Antibodies and their fragments as anti-cancer agents. Curr Pharm Des 12(3): 363-378.
  23. Akhtar S (2006) Non-viral cancer gene therapy: beyond delivery. Gene Ther 13(9): 739-740.
  24. Aiache JM (1991) The ideal drug delivery system: a look into the future. J Aerosol Med 4(4): 323-334.
  25. Wu GY, Wu CH (1987) Receptor-mediated in vitro gene transformation by a soluble DNA carrier system. J Biol Chem 262(10): 4429-4432.
  26. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM (2016) PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 99: 28-51.
  27. Dozier JK, Distefano MD (2015) Site-Specific PEGylation of Therapeutic Proteins. Int J Mol Sci 16(10): 25831-25864.
  28. Joralemon MJ, McRae S, Emrick T (2010) PEGylated polymers for medicine: from conjugation to self-assembled systems. Chem Commun 46(9): 1377-1393.
  29. Rensen PCN, de Vrueh RLA, Kuiper J, Bijsterbosch MK, Biessen EAL, et al. (2001?) Recombinant lipoproteins: lipoprotein-like lipid particles for drug targeting. Adv Drug Deliv Rev 47(2-3): 251-276.
  30. Akbarzadeh A, Rezaei RS, Davaran S, Joo SW, Zarghami N, et al. (2013) Liposome: classification, preparation, and applications. Nanoscale Res Lett 8(1): 102.
  31. Dipak SP, Matthew PK, Sathy VBI (2010) Delivery of therapeutic proteins. J Pharm Sci 99(6): 2557-2575.
  32. Kularatne SA, Wang K, Santhapuram HK, Low PS (2009) Prostate-Specific Membrane Antigen Targeted Imaging and Therapy of Prostate Cancer Using a PSMA Inhibitor as a Homing Ligand. Mol Pharmaceutics 6(3): 780-789.
  33. Trivedi R, Kompella UB (2010) Nanomicellar formulations for sustained drug delivery: strategies and underlying principles. Nanomedicine 5(3): 485-505.
  34. Yuan ZX, He XK, Wu XJ, Gao Y, Fan M, et al. (2014) Peptide fragments of human serum albumin as novel renal targeting carriers. Int J Pharm 460(1-2): 196-204.
  35. Jorgensen JA, Longva AS, Hovig E, Boe SL (2013) Evaluation of biodegradable peptide carriers for light-directed targeting. Nucleic Acid The 23(2): 131-139.
  36. Hamad I, Moghimi SM (2008) Critical issues in site-specific targeting of solid tumours: the carrier, the tumour barriers and the bioavailable drug. Expert Opin Drug Deliv 5(2): 205-219.
  37. Neerman MF (2006) Enhancing the site-specific targeting of macromolecular anticancer drug delivery systems. Curr Drug Targets 7(2): 229-235.
  38. Chin MP, Wrolstad D, Bakris GL, Chertow GM, de Zeeuw D, et al. (2014) Risk factors for heart failure in patients with type 2 diabetes mellitus and stage 4 chronic kidney disease treated with bardoxolone methyl. J Card Fail 20(12): 953-958.
  39. Molema G, Meijer DKF (2001) Drug targeting: organ-specific strategies. Wiley-VCH.
  40. Zhou P, Sun X, Zhang Z (2014) Kidney-targeted drug delivery systems. Acta Pharm Sin B 4(1): 37-42.
  41. Haverdings RFG, Haas M, Greupink AR, deVries PAM, Moolenaar F, et al. (2001) Potentials and limitations of the low-molecular-weight protein lysozyme as a carrier for renal drug targeting. Ren Fail 23(3-4): 397-409.
  42. Haas M, Moolenaar F, Meijer DKF, de Zeeuw D (2002) Specific Drug Delivery to the Kidney. Cardiovasc. Drugs Ther 16(6): 489-496.
  43. Schechter B, Arnon R, Colas C, Burakova T, Wilchek M (1995) Renal accumulation of streptavidin: potential use for targeted therapy to the kidney. Kidney Int 47(5): 1327-1335.
  44. Jang W, Nakagishi Y, Nishiyama N, Kawauchi S, Morimoto Y, et al. (2006) Polyion complex micelles for photodynamic therapy: Incorporation of dendritic photosensitizer excitable at long wavelength relevant to improved tissue-penetrating property. J Control Release 113(1): 73-79.
  45. Kamada H, Tsutsumi Y, Kamada KS, Yamamoto Y, Yoshioka Y, et al. (2003) Synthesis of a poly(vinylpyrrolidone-co-dimethyl maleic anhydride) co-polymer and its application for renal drug targeting. Nat Biotechnol 21(4): 399-404.
  46. Breimer DD (1999) Future challenges for drug delivery. J Control Release 62(1-2): 3-6.
  47. Suzuki K, Susaki H, Okuno S, Sugiyama Y (1999) Renal drug targeting using a vector "alkylglycoside". J Pharmacol Exp Ther 288(1): 57-64.
  48. Shirota K, Kato Y, Suzuki K, Sugiyama Y (2001) Characterization of novel kidney-specific delivery system using an alkylglucoside vector. J Pharmacol Exp Ther 299(2): 459-467.
  49. Haas M, Kluppel AC, Wartna ES, Moolenaar F, Meijer DK, et al. (1997) Drug-targeting to the kidney: renal delivery and degradation of a naproxen-lysozyme conjugate in vivo. Kidney Int 52(6): 1693-1699.
  50. Franssen EJ, Amsterdam RGV, Visser J, Moolenaar F, de Zeeuw D, et al. (1991) Low molecular weight proteins as carriers for renal drug targeting: naproxen-lysozyme. Pharm Res 8(10): 1223-1230.
  51. Franssen EJ, Moolenaar F, de Zeeuw D, Meijer DK (1993) Low molecular weight proteins as carriers for renal drug targeting: naproxen coupled to lysozyme via the spacer L-lactic acid. Pharm Res 10(7): 963-969.
  52. Zhang Z, Zheng Q, Han J, Gao G, Liu J, et al. (2009) The targeting of 14-succinate triptolide-lysozyme conjugate to proximal renal tubular epithelial cells. Biomaterials 30(7): 1372-1381.
  53. Zheng Q, Gong T, Sun X, Zhang ZR (2006) Synthesis, characterization and in vitro evaluation of triptolide-lysozyme conjugate for renal targeting delivery of triptolide. Arch Pharm Res 29(12): 1164-1170.
  54. Prakash J, Weemaes AMVL, Haas M, Proost JH, Meijer DK, et al. (2005) Renal-selective delivery and angiotensin-converting enzyme inhibition by subcutaneously administered captopril-lysozyme. Drug Metab Dispos 33(5): 683-688.
  55. Prakash J, Kok RJ, Weemaes AMVL, Haas M, Proost JH, et al. (2005) Renal targeting of captopril using subcutaneous administration of captopril-lysozyme conjugate. J Control Release 101(1-3): 350-351.
  56. Windt WA, Prakash J, Kok RJ, Moolenaar F, Kluppel CA, et al. (2004) Renal targeting of captopril using captopril-lysozyme conjugate enhances its antiproteinuric effect in adriamycin-induced nephrosis. J Renin Angiotensin Aldosterone Syst 5(4): 197-202.
  57. Kok RJ, Grijpstra F, Walthuis RB, Moolenaar F, de Zeeuw D, et al. (1999) Specific delivery of captopril to the kidney with the prodrug captopril-lysozyme. J Pharmacol Exp Ther 288(1): 281-285.
  58. Dolman ME, Harmsen S, Pieters EH, Sparidans RW, Lacombe M, et al. (2012) Targeting of a platinum-bound sunitinib analog to renal proximal tubular cells. Int J Nanomedicine 7: 417-433.
  59. Dolman ME, Dorenmalen KMV, Pieters EH, Sparidans RW, Lacombe M, et al. (2012) Dendrimer-based macromolecular conjugate for the kidney-directed delivery of a multitargeted sunitinib analogue. Macromol Biosci 12(1): 93-103.
  60. Dolman ME, Harmsen S, Storm G, Hennink WE, Kok RJ (2010) Drug targeting to the kidney: Advances in the active targeting of therapeutics to proximal tubular cells. Adv Drug Deliv Rev 62(14): 1344-1357.
  61. Maack T, Johnson Z, Kau ST, Figueiredo J, Sigulem D (1979) Renal filtration, transport, and metabolism of low-molecular-weight proteins: a review. Kidney Int 16(3): 251-270.
  62. Dreisbach AW, Batuman V (1994) Low-molecular-weight protein competition for binding sites on renal brush border membranes. Ren Physiol Biochem 17(6): 287-293.
  63. Franssen EJ, Koiter J, Kuipers CA, Bruins AP, Moolenaar F, et al. (1992) Low molecular weight proteins as carriers for renal drug targeting. Preparation of drug-protein conjugates and drug-spacer derivatives and their catabolism in renal cortex homogenates and lysosomal lysates. J Med Chem 35(7): 1246-1259.
  64. Cojocel C, Sieveking MF, Beckmann G, Baumann K (1981) Inhibition of renal accumulation of lysozyme (basic low molecular weight protein) by basic proteins and other basic substances. Pflugers Arch 390(3): 211-215.
  65. Yuan ZX, Zhang ZR, Zhu D, Sun X, Gong T, et al. (2009) Specific renal uptake of randomly 50% N-acetylated low molecular weight chitosan. Mol Pharm 6(1): 305-314.
  66. Yuan ZX, Sun X, Gong T, Ding H, Fu Y, et al. (2007) Randomly 50% N-acetylated low molecular weight chitosan as a novel renal targeting carrier. J Drug Target 15(4): 269-278.
  67. He XK, Yuan ZX, Wu XJ, Xu CQ, Li WY (2012) Low molecular weight hydroxyethyl chitosan-prednisolone conjugate for renal targeting therapy: synthesis, characterization and in vivo studies. Theranostics 2(11): 1054-1063.
  68. Yuan ZX, Li JJ, Zhu D, Sun X, Gong T, et al. (2011) Enhanced accumulation of low-molecular-weight chitosan in kidneys: a study on the influence of N-acetylation of chitosan on the renal targeting. J Drug Target 19(7): 540-551.
  69. Christensen EI, Birn H, Verroust P, Moestrup SK (1998) Megalin-mediated endocytosis in renal proximal tubule. Ren Fail 20(2): 191-199.
  70. Liang Z, Gong T, Sun X, Tang JZ, Zhang Z (2012) Chitosan oligomers as drug carriers for renal delivery of zidovudine. Carbohydrate polymers 87: 2284-2290.
  71. Yamamoto Y, Tsutsumi Y, Yoshioka Y, Kamada H, Kamada KS, et al. (2004) Poly(vinylpyrrolidone-co-dimethyl maleic acid) as a novel renal targeting carrier. J Control Release 95(2): 229-237.
  72. Kodaira H, Tsutsumi Y, Yoshioka Y, Kamada H, Kaneda Y, et al. (2004) The targeting of anionized polyvinylpyrrolidone to the renal system. Biomaterials 25(18): 4309-4315.
  73. Vijayakumar S, Peng H, Schwartz GJ (2013) Galectin-3 mediates oligomerization of secreted hensin using its carbohydrate-recognition domain. Am J Physiol Renal Physiol 305(1): F90-F99.
  74. Geng Q, Sun X, Gong T, Zhang ZR (2012) Peptide-drug conjugate linked via a disulfide bond for kidney targeted drug delivery. Bioconjug Chem 23(6): 1200-1210.
  75. Wischnjow A, Sarko D, Janzer M, Kaufman C, Beijer B, et al. (2016) Renal Targeting: Peptide-Based Drug Delivery to Proximal Tubule Cells. Bioconjug Chem 27(4): 1050-1057.
  76. Lin YC, Hung GU, Luo TY, Tsai SC, Sun SS, et al. (2007) Reducing renal uptake of 111In-DOTATOC: a comparison among various basic amino acids. Ann Nucl Med 21(1): 79-83.
  77. Kaunitz JD, Cummins VP, Mishler D, Nagami GT (1993) Inhibition of gentamicin uptake into cultured mouse proximal tubule epithelial cells by L-lysine. J Clin Pharmacol 33(1): 63-69.
  78. Choi CH, Zuckerman JE, Webster P, Davis ME (2011) Targeting kidney mesangium by nanoparticles of defined size. Proc Natl Acad Sci USA 108(16): 6656-6661.
  79. Tuffin G, Waelti E, Huwyler J, Hammer C, Marti HP (2005) Immunoliposome targeting to mesangial cells: a promising strategy for specific drug delivery to the kidney. J Am Soc Nephrol 16(11): 3295-3305.
Creative Commons Attribution License

©2016 Sarko, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.