Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2415 word(s) 2415 2021-08-17 05:56:44

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Duong, V. P-Glycoprotein Inhibitors. Encyclopedia. Available online: https://encyclopedia.pub/entry/13319 (accessed on 28 March 2024).
Duong V. P-Glycoprotein Inhibitors. Encyclopedia. Available at: https://encyclopedia.pub/entry/13319. Accessed March 28, 2024.
Duong, Van-An. "P-Glycoprotein Inhibitors" Encyclopedia, https://encyclopedia.pub/entry/13319 (accessed March 28, 2024).
Duong, V. (2021, August 18). P-Glycoprotein Inhibitors. In Encyclopedia. https://encyclopedia.pub/entry/13319
Duong, Van-An. "P-Glycoprotein Inhibitors." Encyclopedia. Web. 18 August, 2021.
P-Glycoprotein Inhibitors
Edit

P-gp inhibitors are compounds that block or bypass P-gp efflux. The concurrent administration of P-gp inhibitors with P-gp substrates can prevent the expulsion of these substrates and increase their therapeutic effects. Researchers have identified, studied, and evaluated various P-gp inhibitors, including small molecules, natural products, and pharmaceutically inert excipients.

P-gp inhibitors permeability drug delivery solid lipid nanoparticles micelles liposomes polymeric nanoparticles emulsions

1. Introduction

The role of P-gp efflux in the pharmacokinetics of P-gp substrates has been increasingly appreciated [1]. It is well-known that in the human intestine, P-gp is highly expressed in epithelial cells of the colon and ileum (apical side). The expression level in the jejunum, duodenum, and stomach is relatively reduced compared to the ileum [2]. During oral absorption, drug properties (solubility and permeability) and P-gp efflux across the intestinal apical membrane determine the rate and amount of drug diffusing across the basolateral membrane to enter the general circulation [3]. Therefore, P-gp efflux screening is a key step in the drug discovery stage, aided by in vitro, in situ permeability studies and transgenic mdr knockout mice. It has been found that in vitro and in situ P-gp function for a given drug is well correlated with in vivo P-gp activity [4].

To overcome these problems, various P-gp inhibitors have been widely studied. They include small molecule drugs (active pharmaceutical ingredients (APIs) and new chemical entities (NCEs)), natural constituents, and pharmaceutically inert excipients [5][6][7]. In general, P-gp inhibitors can block drug-binding sites competitively, non-competitively, or allosterically, interfere with ATP hydrolysis, and alter the integrity of cell membranes [8]. In particular, for oral drug delivery, these inhibitors improve intestinal absorption, tissue distribution, and reduce substrate metabolism and elimination, resulting in enhanced pharmacokinetic properties and oral bioavailability [8]

Small molecule P-gp inhibitors can be co-administered with P-gp substrates to enhance their oral bioavailability. However, this approach can result in drug-drug interactions and increased side effects because these small molecules have their own pharmacological activities. On the other hand, nonspecific P-gp inhibitors such as polymers, surfactants, and lipid-based excipients can be alternatives to these small molecules. They are safe, pharmaceutically acceptable, and are not absorbed by the gut [9]. They are incorporated in pharmaceutical formulations and indirectly inhibit P-gp by affecting the lipid membrane. Numerous formulations with inherent P-gp inhibitory activity have been studied for many years. These include micelles, emulsions, liposomes, microspheres, solid lipid nanoparticles, polymeric nanoparticles, dendrimers, and solid dispersions [10][11]. In addition to the P-gp inhibitory effect of the excipients, the formulations themselves can improve drug solubility and affinity to the intestinal membrane, promote paracellular passage and endocytic uptake, and enable the lymph transport pathway, resulting in improved oral bioavailability [12][13]

2. Mechanism of P-gp Efflux and Functions of P-gp

P-gp inhibitors are often required to reduce the activity of P-gp. For oral drug administration, inhibiting P-gp can increase drug absorption and bioavailability and thus its therapeutic effects [14]. P-gp inhibition in the BBB can increase the transport of some brain-targeted drugs through the BBB and improve its effects in the brain [15]. In the case of an HIV treatment, P-gp inhibitors are useful to increase drug loading of the fetus shortly before birth, which can reduce the frequency of HIV transmission during birth [16]. P-gp inhibitors can reverse the MDR phenomenon associated with P-gp efflux, resulting in the improved efficiency of chemotherapeutic agents and enhanced pharmacokinetic and pharmacodynamic profiles of drugs.

In oral drug delivery, some models have been used to determine the drug permeability and evaluate the P-gp efflux, such as Caco-2 or Madin Darby canine kidney (MDCK)-MDR1 cell monolayers, inside-out vesicles, single-pass intestinal perfusion (SPIP), and everted gut sac permeability studies. P-gp efflux can be determined by comparing the permeability of a drug between a test sample (a formulation) and a control sample (usually a drug solution) [17][18][19].

3. Classification of P-gp Inhibitors

P-gp inhibitors are compounds that block or bypass P-gp efflux. The concurrent administration of P-gp inhibitors with P-gp substrates can prevent the expulsion of these substrates and increase their therapeutic effects. Researchers have identified, studied, and evaluated various P-gp inhibitors, including small molecules, natural products, and pharmaceutically inert excipients [20]. Additionally, prodrugs, synthetic peptides, and P-gp expression suppressors are also potential approaches to overcome P-gp efflux [21].

A summary of the P-gp inhibitors is presented in Table 1.

Table 1. Classification of P-glycoprotein inhibitors.

Classification Examples
Small molecules First generation Verapamil, cyclosporine, trifluoperazine, quinidine, reserpine, yohimbine, tamoxifen, toremifene, and vincristine
Second generation Dexverapamil and PSC 833 (valspodar)
Third generation VX-710 (biricodar), GF120918 (elacridar), LY335979 (zosuquidar), XR9576 (tariquidar), R101933 (laniquidar), WK-X-34, and OC144-093 (ontogeny)
Natural products Alkaloids Glaucine, pervilleine, berberine, kopsiflorine, lobeline, cepharanthine, ibogaine, theobromine
Flavonoids Quercetin, morin, phloretin, rhamnetin, plagiochin E, daidzin, procyanidine, rotenone
Coumarins Decursinol, bergaptol, galbanic acid, farnesiferol
Terpenoid Citral, latilagascene, paraliane, pepluanin A, jolkinol B, euphoportlandol lhelioscopinolide, tuckeyanols, euphotuckeyanol, isopimaric acid, totarol
Saponins Gracillin, tenacissimoside A, karavilagenin C, balsaminol, ginsenoside F1, protopanaxatriol
Peptides Discodermolide, kendarimide, hapalosin, nocardioazine
Resins Gambogic acid, orizabin
Miscellaneous natural compounds Acetoxy cavicolacetate, arctigenin, pheophorbide, porphyrin, cannabinol, gomisin, pregomisin, phenylbutanoid
Pharmaceutical excipients Surfactants Polysorbates: polysorbate 80, 20
Sucrose esters: sucrose monolaurate
Tocopheryl ester: TPGS
PEG esters: Cremophor EL, Solutol HS-15, Labrasol, Softigen 767, Myrj 52, Gelucire 44/14
PEG ethers: Brij 78
Other: sodium 1,4-bis (2-ethylhexoxy)-1,4-dioxobutane-2-sulfonate (AOT), cetyltrime-thylammonium bromide (CTAB)
Polymers Natural polymers: dextrans, agar, gellan gum, gum arabic, gum traganth, guar gum, carrageenan gum, xanthan gum, alginates, chitosan
Amphiphilic diblock copolymers: methoxyPEG-block-polycaprolactone (MePEG-b-PCL), Soluplus
Pluronic block copolymers: poloxamer 407 and 188
Others Glycerides: monoolein (PeceolTM), monostearin
Phospholipids: 8:0 PC, 10:0 PC
Methylated cyclodextrin

TPGS: α-tocopheryl polyethylene glycol-1000-succinate, PEG: polyethylene glycol.

4. Recently Developed Formulations to Bypass P-gp and Enhance Oral Bioavailability of P-gp Substrates

In addition to P-gp inhibitory effects due to the presence of P-gp inhibitors in their components, which result in increased intestinal absorption and bioavailability of P-gp substrates, these drug delivery systems, particularly nanocarriers, can also enhance oral bioavailability by various effects [22]. (1) They increase drug solubility, dissolution, and affinity to the intestinal membrane, leading to an increase in drug absorption by enterocytes [5]. Most drug delivery systems can also protect drugs from degradation in the gastrointestinal tract [23]. (2) Some formulations can improve mucoadhesion due to the interaction between the nanocarrier (positive charge) and mucin (negative charge) [24]. (3) Nanocarriers may interact with tight junction proteins and modulate tight junctions in the intestinal epithelium, resulting in paracellular transport [25]. (4) Receptor-mediated endocytosis and the transcytosis of enterocytes can enhance the transcellular transport of nanocarriers [26]. Particles <500 nm in size can be absorbed by intestinal enterocytes and bypass P-gp efflux, making this route an essential transport pathway for nanocarriers [27]. The endocytosis mechanisms of drug delivery systems can be explored by using the everted intestinal ring model. (5) Some nanocarriers, such as SLNs, NLCs [22], polymer–lipid hybrid nanoparticles (PLHNs) [28], and micelles [27], can be phagocytosed by microfold cells (M cells). These are specialized cells in the lymphoid tissue of Peyer’s patches in the small intestine. In other parts of the gastrointestinal tract, M cells are located in the mucosa-associated lymphoid tissue. They allow the transport of microbes and small particles across the epithelial cell layer. After the selective endocytosis of antigens, M cells transport them to intraepithelial macrophages and lymphocytes prior to their migration to lymph nodes [29]. Thus, the drug can bypass the P-gp efflux and liver metabolism. (6) Another effect that occurs with various lipid-based drug delivery systems is lymphatic absorption from enterocytes (mediated by lipase and chylomicron uptake). It is one of the major absorption mechanisms for drugs encapsulated in SLNs, NLCs, emulsions, and liposomes [30][31]. Lipase digests triglycerides in the stomach to form crude emulsified lipid systems. They are subsequently exposed to bile salts, cholesterol, and pancreatic lipase and further digested to form mixed micelles of the lipidic system containing drugs. Then, these micelles are absorbed by enterocytes and converted to chylomicrons, which can be transported via the mesenteric lymph to enter the lymphatic transport system. The chylomicrons ultimately enter the systemic circulation via lymphatic drainage at the thoracic duct [32].

Different formulations have been developed and employed to enhance the oral absorption and bioavailability of various P-gp substrates. The following sections describe the recent development of pharmaceutical formulations for the oral delivery of a wide range of P-gp substrates.

4.1. Emulsion, Self-Micro, and Nano Emulsifying Drug Delivery Systems

Emulsions are colloidal systems prepared from two immiscible liquids with force and the addition of surfactants, in which one phase is dispersed as tiny droplets in the other phase [33]. The dispersed phase is also called the discontinuous phase or internal phase. The outer phase is also termed as the continuous phase, external phase, or dispersion medium. The surfactants have affinities for both phases and are located on the interfacial surface of the two liquids to stabilize the emulsions. Two types of emulsions are oil-in-water and water-in-oil emulsions, depending on the volume ratio of the two liquids. Methods used to prepare emulsions include ultrasonication, high-pressure homogenization, microfluidization, phase inversion temperature, and spontaneous emulsification [34]. SMEDDS and SNEDDS are lipid-based nanocarriers that spontaneously emulsify to form fine oil-in-water emulsions after exposure to gastrointestinal fluids. SMEDDS and SNEDDS are stable mixtures of oil, surfactants, and cosurfactants. SMEDDS contain a lower percentage of lipids and a higher percentage of hydrophilic surfactants and cosurfactants compared with SNEDDS. SMEDDS usually form thermodynamically stable emulsions <100 nm in size. Emulsions derived from SNEDDS range in size from 100 to 250 nm. Emulsions, SMEDDS, and SNEDDS have been widely used as drug delivery systems to enhance the oral absorption and bioavailability of various lipophilic drugs [35]. These benefits are attributed to the endocytosis of enterocytes, paracellular transport, and lymphatic absorption via chylomicron uptake. In addition, the lipids and surfactants used for the preparation of emulsions, SMEDDS, and SNEDDS may have P-gp inhibitory effects, which considerably reduce P-gp efflux and increase the absorption of P-gp substrates.

4.2. Liposomes

Liposomes are spherical vesicles composed of bilayer membranes, such as those formed by phospholipids. They can incorporate hydrophilic drugs into the water-soluble central compartment and hydrophobic drugs into the bilayer membrane [36]. Liposomes can enhance oral absorption of hydrophobic drugs through several mechanisms that include mucoadhesion, translocation across mucus layers, improved permeation across the enteric epithelia, endocytosis, uptake by M cells, and lymphatic absorption via chylomicron uptake [37]. In addition, phospholipids and other components in liposomes can inhibit P-gp to further improve the permeability of P-gp substrates. However, liposomes are unstable in the harsh gastrointestinal environment because phospholipids aggregate at low pH and with the presence of enzymes, such as pancreatic lipase. Therefore, liposomes are modified by PEGylation, mucin coating, and polymer coating [22]. Some P-gp substrates have been encapsulated in liposomes to improve drug absorption and oral bioavailability.

4.3. SLNs and NLCs

SLNs and NLCs are lipid-based nanoparticles with a solid matrix, which have emerged as alternatives to emulsions, liposomes, and polymeric nanoparticles [38]. SLNs are prepared from solid lipids, surfactants, and co-surfactants. In contrast, in NLCs, liquid oils are added to create imperfect structures that can accommodate more drug [39]. NLCs can increase drug loadings and reduce drug expulsion during storage compared to SLNs. SLNs and NLCs are produced from physiological and biodegradable lipids and other generally recognized as safe (GRAS) materials. Therefore, they are both safe and biocompatible. They can also increase the solubility and stability of drugs encapsulated in solid matrices. SLNs and NLCs can be prepared by various methods that include high-pressure homogenization, emulsion/solvent evaporation, microemulsion, phase inversion, and solvent injection [40][41][42]. SLNs and NLCs can enhance drug absorption and the bioavailability of hydrophilic drugs through several mechanisms, such as uptake by M cells [22], lymphatic absorption from enterocytes via chylomicron uptake [30], paracellular transport through tight junction opening [43], and receptor-mediated endocytosis and transcytosis of enterocytes [26]. In the case of P-gp substrates, lipids and surfactants in SLNs and NLCs could also inhibit P-gp efflux and increase drug permeation [44]

4.4. Micelles

Micelles are spherical amphiphilic structures formed by the self-assembly of amphiphilic molecules. They have a hydrophobic core that is suitable for the encapsulation of poorly water-soluble drugs. A hydrophilic shell can stabilize the hydrophobic core and make the micelles water-soluble [45]. Micelles are nanosized colloidal dispersions with a diameter of 10–200 nm, which avoids elimination by the reticuloendothelial system and increases their circulation time [46]. Polymeric micelles are produced by increasing the polymer concentration above the corresponding CMC. Micelles have the advantages of easy drug encapsulation and easy surface manipulation. Drugs can be encapsulated by chemical covalent attachments or physical methods. In chemical methods, the drugs are covalently cross-linked with polymers, which considerably improves circulation kinetics, biodistribution, and the accumulation of micelles at target sites. However, cross-linking chemical reactions can be challenging and complicated. However, physical methods are practical and simpler. Several widely used techniques include solvent evaporation, oil-in-water emulsion, direct dissolution, dialysis, and freeze-drying [47]. Many surfactants with P-gp inhibitory activity can be used for the preparation of micelles. These include Tween 80, TPGS, Cremophor EL, and Pluronic 85. They can effectively improve the intestinal permeability of P-gp substrates.

4.5. Polymeric Nanoparticles (NPs)

Polymeric NPs are nano-colloidal systems produced from different polymers. They have been widely used for the oral delivery of various chemotherapeutic agents. Polymeric NPs have the advantages of high stability in the gastrointestinal tract and the ability to encapsulate a variety of drug molecules [48]. Polymers used for the preparation of polymeric NPs include natural polymers, such as chitosan, gelatin, dextran, albumin, and alginate, and synthetic biodegradable polymers that include polylactic acid, polyglycolic acid, polylactic and polyglycolic acid copolymers (PLGA), polyethylene imine (PEI), polyalkyl cyanoacrylate (PACA), and PCL [49]. Some polymeric NPs, such as chitosan, typically have mucoadhesive properties that increase their residence time and diffusion in the mucus. They are mainly absorbed in the gastrointestinal tract via endocytosis, such as clathrin-dependent endocytosis for various chitosan and PLGA-NPs. Phagocytosis by M cells is also involved in the absorption of polymeric NPs via lymphatic organs [50]. In addition, some components in polymeric NPs can open tight junctions to increase the paracellular transport of drugs, such as chitosan and its derivatives [48]. Many P-gp substrates have been encapsulated into polymeric micelles to bypass P-gp efflux, thereby enhancing oral bioavailability.

4.6. Other Pharmaceutical Formations for Inhibition of P-gp

In addition to the drug delivery system discussed above, other pharmaceutical formulations can also inhibit or bypass P-gp efflux and enhance drug absorption and oral bioavailability. These include microspheres, solid dispersions, and dendrimers. Microspheres are spherical microparticles 1–1000 μm in size that incorporate drugs within their core. Solid dispersions are dispersions of drugs in solid matrices of polymers or small molecules. The dispersed state can be eutectic mixtures, amorphous/crystalline suspensions, or crystalline/glass solutions [51]. Dendrimers are tree-like branched structures that are used in various targeted drug delivery systems. They consist of an initial core, interior layers with repeating units, and an exterior with terminal functionality [52].

Features of the pharmaceutical formulations in 4.1-4.6 are summarized in Tables 2-6 of the articles.

References

  1. Jiunn H. Lin; Masayo Yamazaki; Role of P-Glycoprotein in Pharmacokinetics. Clinical Pharmacokinetics 2003, 42, 59-98, 10.2165/00003088-200342010-00003.
  2. Mikihisa Takano; Ryoko Yumoto; Teruo Murakami; Expression and function of efflux drug transporters in the intestine. Pharmacology & Therapeutics 2006, 109, 137-161, 10.1016/j.pharmthera.2005.06.005.
  3. Famida G. Hoosain; Yahya Choonara; Lomas K. Tomar; Pradeep Kumar; Charu Tyagi; Lisa du Toit; Viness Pillay; Bypassing P-Glycoprotein Drug Efflux Mechanisms: Possible Applications in Pharmacoresistant Schizophrenia Therapy. BioMed Research International 2015, 2015, 1-21, 10.1155/2015/484963.
  4. Yasuhisa Adachi; Hiroshi Suzuki; Yuichi Sugiyama; Quantitative evaluation of the function of small intestinal P-glycoprotein: comparative studies between in situ and in vitro.. Pharmaceutical Research 2003, 20, 1163-1169, 10.1023/a:1025088628787.
  5. Ting Zhao; Hongli Zhou; Wanyan Wu; Xu Song; Tao Gong; A multistage oral delivery system of PTX for improving oral bioavailability and enhancing anticancer efficacy. Drug Development and Industrial Pharmacy 2021, 47, 259-267, 10.1080/03639045.2021.1879831.
  6. Marcello Leopoldo; Patrizia Nardulli; Marialessandra Contino; Francesco Leonetti; Gert Luurtsema; Nicola Antonio Colabufo; An updated patent review on P-glycoprotein inhibitors (2011-2018). Expert Opinion on Therapeutic Patents 2019, 29, 455-461, 10.1080/13543776.2019.1618273.
  7. Xiaobo Yang; Kexin Liu; P-gp Inhibition-Based Strategies for Modulating Pharmacokinetics of Anticancer Drugs: An Update. Current Drug Metabolism 2016, 17, 806-826, 10.2174/1389200217666160629112717.
  8. Manthena V.S Varma; Yasvanth Ashokraj; Chinmoy S Dey; Ramesh Panchagnula; P-glycoprotein inhibitors and their screening: a perspective from bioavailability enhancement. Pharmacological Research 2003, 48, 347-359, 10.1016/s1043-6618(03)00158-0.
  9. Panayiotis P. Constantinides; Kishor M. Wasan; Lipid Formulation Strategies for Enhancing Intestinal Transport and Absorption of P-Glycoprotein (P-gp) Substrate Drugs: In vitro/In vivo Case Studies. Journal of Pharmaceutical Sciences 2007, 96, 235-248, 10.1002/jps.20780.
  10. Siddharth S. Kesharwani; Shamandeep Kaur; Hemachand Tummala; Abhay T. Sangamwar; Overcoming multiple drug resistance in cancer using polymeric micelles. Expert Opinion on Drug Delivery 2018, 15, 1127-1142, 10.1080/17425247.2018.1537261.
  11. Hossam Abdallah; Ahmed Al-Abd; Riham Salah El-Dine; Ali M. El-Halawany; P-glycoprotein inhibitors of natural origin as potential tumor chemo-sensitizers: A review. Journal of Advanced Research 2014, 6, 45-62, 10.1016/j.jare.2014.11.008.
  12. Pranav Shah; Kejal Chavda; Bhavin Vyas; Shailaja Patel; Formulation development of linagliptin solid lipid nanoparticles for oral bioavailability enhancement: role of P-gp inhibition. Drug Delivery and Translational Research 2020, 11, 1166-1185, 10.1007/s13346-020-00839-9.
  13. Jiang Ni; Fengchun Tian; Fatima Zohra Dahmani; Hui Yang; Deren Yue; Shuwang He; Jianping Zhou; Jing Yao; Curcumin-carboxymethyl chitosan (CNC) conjugate and CNC/LHR mixed polymeric micelles as new approaches to improve the oral absorption of P-gp substrate drugs. Drug Delivery 2016, 23, 3424-3435, 10.1080/10717544.2016.1189625.
  14. Bashaier Mohammed Al-Kandari; Monerah H. Al-Soraj; Mohsen A. Hedaya; Dual Formulation and Interaction Strategies to Enhance the Oral Bioavailability of Paclitaxel. Journal of Pharmaceutical Sciences 2020, 109, 3386-3393, 10.1016/j.xphs.2020.07.027.
  15. Jintao Wang; Yun Shi; Shuchun Yu; Yan Wang; Qingcheng Meng; Ge Liang; Maryellen F. Eckenhoff; Huafeng Wei; Intranasal administration of dantrolene increased brain concentration and duration. PLOS ONE 2020, 15, e0229156, 10.1371/journal.pone.0229156.
  16. Alfred H. Schinkel; Johan Jonker; Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview. Advanced Drug Delivery Reviews 2012, 64, 138-153, 10.1016/j.addr.2012.09.027.
  17. Han-Joo Maeng; Ho-Jung Yoo; In-Wha Kim; Im-Sook Song; Suk-Jae Chung; Chang-Koo Shim; P-Glycoprotein–Mediated Transport of Berberine across Caco-2 Cell Monolayers. Journal of Pharmaceutical Sciences 2002, 91, 2614-2621, 10.1002/jps.10268.
  18. Jianghong Fan; Han-Joo Maeng; K. Sandy Pang; Interplay of transporters and enzymes in the Caco-2 cell monolayer: I. effect of altered apical secretion. Biopharmaceutics & Drug Disposition 2010, 31, 215-227, 10.1002/bdd.704.
  19. Jin-Wook Kang; Hyun-Jong Cho; Hyo Jung Lee; Hyo-Eon Jin; Han-Joo Maeng; Polyethylene glycol-decorated doxorubicin/carboxymethyl chitosan/gold nanocomplex for reducing drug efflux in cancer cells and extending circulation in blood stream. International Journal of Biological Macromolecules 2018, 125, 61-71, 10.1016/j.ijbiomac.2018.12.028.
  20. Naseem Akhtar; Abdul Ahad; Roop Kishan Khar; Manu Jaggi; Mohd Aqil; Zeenat Iqbal; Farhan Ahmad; Sushama Talegaonkar; The emerging role of P-glycoprotein inhibitors in drug delivery: a patent review. Expert Opinion on Therapeutic Patents 2011, 21, 561-576, 10.1517/13543776.2011.561784.
  21. Xun Li; Hongyu Yuan; Jifeng Wu; Jinpei Li; Xianjun Qu; Wenfang Xu; Wei Tang; Strategies to overcome or circumvent P-glycoprotein mediated multidrug resistance.. Current Medicinal Chemistry 2008, 15, 470-476, 10.2174/092986708783503258.
  22. Kaushik Thanki; Rahul P. Gangwal; Abhay T. Sangamwar; Sanyog Jain; Oral delivery of anticancer drugs: Challenges and opportunities. Journal of Controlled Release 2013, 170, 15-40, 10.1016/j.jconrel.2013.04.020.
  23. Silvia Mazzaferro; Kawthar Bouchemal; Gilles Ponchel; Oral delivery of anticancer drugs III: formulation using drug delivery systems. Drug Discovery Today 2013, 18, 99-104, 10.1016/j.drudis.2012.08.007.
  24. Tian'e Chen; Liangxing Tu; Ge Wang; Na Qi; Wei Wu; Wei Zhang; Jianfang Feng; Multi-functional chitosan polymeric micelles as oral paclitaxel delivery systems for enhanced bioavailability and anti-tumor efficacy. International Journal of Pharmaceutics 2020, 578, 119105, 10.1016/j.ijpharm.2020.119105.
  25. Mária A. Deli; Potential use of tight junction modulators to reversibly open membranous barriers and improve drug delivery. Biochimica et Biophysica Acta (BBA) - Biomembranes 2009, 1788, 892-910, 10.1016/j.bbamem.2008.09.016.
  26. E. Roger; F. Lagarce; E. Garcion; J.-P. Benoit; Lipid nanocarriers improve paclitaxel transport throughout human intestinal epithelial cells by using vesicle-mediated transcytosis. Journal of Controlled Release 2009, 140, 174-181, 10.1016/j.jconrel.2009.08.010.
  27. Parvin Zakeri-Milani; Badir Delf Loveymi; Mitra Jelvehgari; Hadi Valizadeh; The characteristics and improved intestinal permeability of vancomycin PLGA-nanoparticles as colloidal drug delivery system. Colloids and Surfaces B: Biointerfaces 2013, 103, 174-181, 10.1016/j.colsurfb.2012.10.021.
  28. Sanyog Jain; Pankaj U. Valvi; Nitin K. Swarnakar; Kaushik Thanki; Gelatin Coated Hybrid Lipid Nanoparticles for Oral Delivery of Amphotericin B. Molecular Pharmaceutics 2012, 9, 2542-2553, 10.1021/mp300320d.
  29. N A Mabbott; David Donaldson; Hiroshi Ohno; I R Williams; A Mahajan; Microfold (M) cells: important immunosurveillance posts in the intestinal epithelium. Mucosal Immunology 2013, 6, 666-677, 10.1038/mi.2013.30.
  30. Natalie Trevaskis; William Charman; Christopher Porter; Lipid-based delivery systems and intestinal lymphatic drug transport: A mechanistic update. Advanced Drug Delivery Reviews 2008, 60, 702-716, 10.1016/j.addr.2007.09.007.
  31. Shailesh S. Chalikwar; Veena S. Belgamwar; Vivek R. Talele; Sanjay J. Surana; Mrunal U. Patil; Formulation and evaluation of Nimodipine-loaded solid lipid nanoparticles delivered via lymphatic transport system. Colloids and Surfaces B: Biointerfaces 2012, 97, 109-116, 10.1016/j.colsurfb.2012.04.027.
  32. Christopher Porter; William N. Charman; Uptake of drugs into the intestinal lymphatics after oral administration. Advanced Drug Delivery Reviews 1997, 25, 71-89, 10.1016/s0169-409x(96)00492-9.
  33. Asmaa Elzayat; Inés Adam-Cervera; Olaia Álvarez-Bermúdez; Rafael Muñoz-Espí; Nanoemulsions for synthesis of biomedical nanocarriers. Colloids and Surfaces B: Biointerfaces 2021, 203, 111764, 10.1016/j.colsurfb.2021.111764.
  34. Manjit Jaiswal; Rupesh Dudhe; Pawan K Sharma; Nanoemulsion: an advanced mode of drug delivery system. 3 Biotech 2014, 5, 123-127, 10.1007/s13205-014-0214-0.
  35. Suphatra Hiranphinyophat; Akihisa Otaka; Yuta Asaumi; Syuji Fujii; Yasuhiko Iwasaki; Particle-stabilized oil-in-water emulsions as a platform for topical lipophilic drug delivery. Colloids and Surfaces B: Biointerfaces 2020, 197, 111423, 10.1016/j.colsurfb.2020.111423.
  36. Vladimir P. Torchilin; Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery 2005, 4, 145-160, 10.1038/nrd1632.
  37. Haisheng He; Yi Lu; Jianping Qi; Quangang Zhu; Zhongjian Chen; Wei Wu; Adapting liposomes for oral drug delivery. Acta Pharmaceutica Sinica B 2018, 9, 36-48, 10.1016/j.apsb.2018.06.005.
  38. R.H. Müller; M. Radtke; S.A. Wissing; Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. Advanced Drug Delivery Reviews 2002, 54, S131-S155, 10.1016/s0169-409x(02)00118-7.
  39. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Sang-Cheol Chi; Preparation of Ondansetron Hydrochloride-Loaded Nanostructured Lipid Carriers Using Solvent Injection Method for Enhancement of Pharmacokinetic Properties. Pharmaceutical Research 2019, 36, 1-12, 10.1007/s11095-019-2672-x.
  40. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Preparation of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers for Drug Delivery and the Effects of Preparation Parameters of Solvent Injection Method. Molecules 2020, 25, 4781, 10.3390/molecules25204781.
  41. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Sang-Cheol Chi; Nanostructured lipid carriers containing ondansetron hydrochloride by cold high-pressure homogenization method: Preparation, characterization, and pharmacokinetic evaluation. Journal of Drug Delivery Science and Technology 2019, 53, 101185, 10.1016/j.jddst.2019.101185.
  42. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Sang-Cheol Chi; Data on optimization and drug release kinetics of nanostructured lipid carriers containing ondansetron hydrochloride prepared by cold high-pressure homogenization method. Data in Brief 2019, 26, 104475, 10.1016/j.dib.2019.104475.
  43. Mingfei Yao; David McClements; Hang Xiao; Improving oral bioavailability of nutraceuticals by engineered nanoparticle-based delivery systems. Current Opinion in Food Science 2014, 2, 14-19, 10.1016/j.cofs.2014.12.005.
  44. Ander Estella-Hermoso de Mendoza; Veronique Préat; Faustino Mollinedo; Maria J Blanco-Prieto; In vitro and in vivo efficacy of edelfosine-loaded lipid nanoparticles against glioma. Journal of Controlled Release 2011, 156, 421-426, 10.1016/j.jconrel.2011.07.030.
  45. Maximiliano Cagel; Fiorella C. Tesan; Ezequiel Bernabeu; Maria J. Salgueiro; Marcela B. Zubillaga; Marcela A. Moretton; Diego A. Chiappetta; Polymeric mixed micelles as nanomedicines: Achievements and perspectives. European Journal of Pharmaceutics and Biopharmaceutics 2017, 113, 211-228, 10.1016/j.ejpb.2016.12.019.
  46. Loujin Houdaihed; James C. Evans; Christine Allen; Overcoming the Road Blocks: Advancement of Block Copolymer Micelles for Cancer Therapy in the Clinic. Molecular Pharmaceutics 2017, 14, 2503-2517, 10.1021/acs.molpharmaceut.7b00188.
  47. Abhirup Mandal; Rohit Bisht; Ilva D. Rupenthal; Ashim K. Mitra; Polymeric micelles for ocular drug delivery: From structural frameworks to recent preclinical studies. Journal of Controlled Release 2017, 248, 96-116, 10.1016/j.jconrel.2017.01.012.
  48. Laurence Plapied; Nicolas Duhem; Anne Des Rieux; Véronique Préat; Fate of polymeric nanocarriers for oral drug delivery. Current Opinion in Colloid & Interface Science 2011, 16, 228-237, 10.1016/j.cocis.2010.12.005.
  49. Avnesh Kumari; Sudesh Kumar Yadav; Subhash C. Yadav; Biodegradable polymeric nanoparticles based drug delivery systems. Colloids and Surfaces B: Biointerfaces 2010, 75, 1-18, 10.1016/j.colsurfb.2009.09.001.
  50. Gaurav Sahay; Daria Y. Alakhova; Alexander V. Kabanov; Endocytosis of nanomedicines. Journal of Controlled Release 2010, 145, 182-195, 10.1016/j.jconrel.2010.01.036.
  51. Yanbin Huang; Wei-Guo Dai; Fundamental aspects of solid dispersion technology for poorly soluble drugs. Acta Pharmaceutica Sinica B 2013, 4, 18-25, 10.1016/j.apsb.2013.11.001.
  52. Serge Mignani; Xangyang Shi; João Rodrigues; Helena Tomas; Andrii Karpus; Jean-Pierre Majoral; First-in-class and best-in-class dendrimer nanoplatforms from concept to clinic: Lessons learned moving forward. European Journal of Medicinal Chemistry 2021, 219, 113456, 10.1016/j.ejmech.2021.113456.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 800
Revision: 1 time (View History)
Update Date: 18 Aug 2021
1000/1000