Please wait a minute...
Frontiers of Chemical Science and Engineering

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

Postal Subscription Code 80-969

2018 Impact Factor: 2.809

Front Chem Sci Eng    2013, Vol. 7 Issue (1) : 9-19    https://doi.org/10.1007/s11705-013-1306-9
REVIEW ARTICLE
Overcoming oral insulin delivery barriers: application of cell penetrating peptide and silica-based nanoporous composites
Huining HE1,4,5, Junxiao YE1, Jianyong SHENG2, Jianxin WANG2, Yongzhuo HUANG2,3, Guanyi CHEN4, Jingkang WANG1(), Victor C YANG5,6()
1. School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; 2. Department of Pharmaceutics, School of Pharmacy, Fudan University; Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Shanghai 201203, China; 3. Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; 4. School of Environmental Science and Engineering, State Key Laboratory of Engines, Tianjin University, Tianjin 300072, China; 5. Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; 6. Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Michigan 48109-1065, USA
 Download: PDF(551 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Oral insulin delivery has received the most attention in insulin formulations due to its high patient compliance and, more importantly, to its potential to mimic the physiologic insulin secretion seen in non-diabetic individuals. However, oral insulin delivery has two major limitations: the enzymatic barrier that leads to rapid insulin degradation, and the mucosal barrier that limits insulin’s bioavailability. Several approaches have been actively pursued to circumvent the enzyme barrier, with some of them receiving promising results. Yet, thus far there has been no major success in overcoming the mucosal barrier, which is the main cause in undercutting insulin’s oral bioavailability. In this review of our group’s research, an innovative silica-based, mucoadhesive oral insulin formulation with encapsulated-insulin/cell penetrating peptide (CPP) to overcome both enzyme and mucosal barriers is discussed, and the preliminary and convincing results to confirm the plausibility of this oral insulin delivery system are reviewed. In vitro studies demonstrated that the CPP-insulin conjugates could facilitate cellular uptake of insulin while keeping insulin’s biologic functions intact. It was also confirmed that low molecular weight protamine (LMWP) behaves like a CPP peptide, with a cell translocation potency equivalent to that of the widely studied TAT. The mucoadhesive properties of the produced silica-chitosan composites could be controlled by varying both the pH and composition; the composite consisting of chitosan (25 wt-%) and silica (75 wt-%) exhibited the greatest mucoadhesion at gastric pH. Furthermore, drug release from the composite network could also be regulated by altering the chitosan content. Overall, the universal applicability of those technologies could lead to development of a generic platform for oral delivery of many other bioactive compounds, especially for peptide or protein drugs which inevitably encounter the poor bioavailability issues.

Keywords insulin      cell penetrating peptide      mucoadhesive composites      oral delivery     
Corresponding Author(s): WANG Jingkang,Email:wangjkch@tju.edu.cn; YANG Victor C,Email:vcyang@umich.edu   
Issue Date: 05 March 2013
 Cite this article:   
Huining HE,Junxiao YE,Jianyong SHENG, et al. Overcoming oral insulin delivery barriers: application of cell penetrating peptide and silica-based nanoporous composites[J]. Front Chem Sci Eng, 2013, 7(1): 9-19.
 URL:  
https://academic.hep.com.cn/fcse/EN/10.1007/s11705-013-1306-9
https://academic.hep.com.cn/fcse/EN/Y2013/V7/I1/9
Fig.1  Schematic diagrams of (a) silica/alginate-based mucoadhesive nanocomposite loaded with insulin-CPP conjugates, (b) mechanism of action of the proposed oral insulin delivery system in allowing insulin to target intestinal mucosa and cross through the epithelial layer (Reprinting with permission)
Fig.1  Schematic diagrams of (a) silica/alginate-based mucoadhesive nanocomposite loaded with insulin-CPP conjugates, (b) mechanism of action of the proposed oral insulin delivery system in allowing insulin to target intestinal mucosa and cross through the epithelial layer (Reprinting with permission)
Fig.2  Synthesis and characterization of the insulin-TAT conjugates. (a) Insulin was linked to TAT using SMCC as a crosslinker; (b) SDS-PAGE analysis of protein bands either stained with Coomassie brilliant blue (left panel) or visualized with UV light (right 2 panels) prior to HPLC (left two panels) and after HPLC (right panel) purification, where lane 1 is free insulin and lane 2 is insulin-TAT conjugates; (c) elution profile of insulin, insulin-TAT conjugates, and TAT using a heparin affinity column (Reprinting with permission)
Fig.2  Synthesis and characterization of the insulin-TAT conjugates. (a) Insulin was linked to TAT using SMCC as a crosslinker; (b) SDS-PAGE analysis of protein bands either stained with Coomassie brilliant blue (left panel) or visualized with UV light (right 2 panels) prior to HPLC (left two panels) and after HPLC (right panel) purification, where lane 1 is free insulin and lane 2 is insulin-TAT conjugates; (c) elution profile of insulin, insulin-TAT conjugates, and TAT using a heparin affinity column (Reprinting with permission)
Fig.3  Fluorescence microphotographs after exposure of Caco-2 cells to (a) FITC-labeled free insulin or (b) insulin-TAT conjugates. (c) In vitro intestinal absorption of the insulin-TAT conjugates. Both apical-to-basal and basal-to-apical absorption by Caco-2 cell monolayers of FITC-labeled insulin and insulin-TAT conjugates were examined. Samples were collected every 2 h and analyzed by fluorescence spectroscopy (Reprinting with permission)
Fig.3  Fluorescence microphotographs after exposure of Caco-2 cells to (a) FITC-labeled free insulin or (b) insulin-TAT conjugates. (c) In vitro intestinal absorption of the insulin-TAT conjugates. Both apical-to-basal and basal-to-apical absorption by Caco-2 cell monolayers of FITC-labeled insulin and insulin-TAT conjugates were examined. Samples were collected every 2 h and analyzed by fluorescence spectroscopy (Reprinting with permission)
Fig.4  Integrity of insulin after translocation of insulin-TAT conjugates. The results from ELISA were plotted against the results from fluorescent assay. Intact (integrity) and enzyme degraded (degradation) insulin-TAT were used as negative and positive controls, respectively. Two samples (1 and 2) were collected from the receiver chamber of the transwell culture plates (Reprinting with permission)
Fig.4  Integrity of insulin after translocation of insulin-TAT conjugates. The results from ELISA were plotted against the results from fluorescent assay. Intact (integrity) and enzyme degraded (degradation) insulin-TAT were used as negative and positive controls, respectively. Two samples (1 and 2) were collected from the receiver chamber of the transwell culture plates (Reprinting with permission)
Amino acid sequence
HIV-TAT47-57GRKKRRQRRRPPQ
LMWPVSRRRRRRGGRRRR
Tab.1  Amino acid sequences for TAT and LMWP
Fig.5  Cellular translocation of LMWP-FITC conjugates into cultured HeLa cells after 30 min of in vitro incubation (left: differential interference contrast image; right: fluorescence microscopy) (Reprinting with permission)
Fig.5  Cellular translocation of LMWP-FITC conjugates into cultured HeLa cells after 30 min of in vitro incubation (left: differential interference contrast image; right: fluorescence microscopy) (Reprinting with permission)
Fig.6  (a) Cellular localization of rhodamine-labeled phalloidin-LMWP conjugates into MG63 osteoblast cells; (b) flow cytometric analysis of AlexFluor-488 labeled phalloidin, LMWP-phalloidin, and TAT-phalloidin in MG63 osteoblast cells after incubation at 37°C for 30 min in a medium containing 10% serum (Reprinting with permission)
Fig.6  (a) Cellular localization of rhodamine-labeled phalloidin-LMWP conjugates into MG63 osteoblast cells; (b) flow cytometric analysis of AlexFluor-488 labeled phalloidin, LMWP-phalloidin, and TAT-phalloidin in MG63 osteoblast cells after incubation at 37°C for 30 min in a medium containing 10% serum (Reprinting with permission)
Fig.7  (a) Tumor regression after intra-tumoral injection of LMWP-gelonin or TAT-gelonin conjugates. Tumor volumes were measured 30 days after cessation of drug treatment. Right: top to bottom represent tumors excised from mice treated with PBS solution (average tumor mass: 3.16±0.65 g); 100 μg gelonin (2.62±0.53 g); 110 μg LMWP-gelonin (0.33±0.12 g); 10 μg LMWP physically mixed with 100 μg gelonin (2.74±0.68 g); and 110 μg TAT-gelonin (0.21±0.04 g). (b) Tumor penetration of rhodamine-labeled free gelonin (top) or LWMP-gelonin conjugates (bottom) into the subcutaneously implanted tumor of a mouse. Ten hours after injection of the labeled drug or drug-conjugates, tumors were excised, processed into cryosections and examined by confocal microscopy. Left and right panels represent fluorescence microphotographs and DIC images, respectively (Reprinting with permission)
Fig.7  (a) Tumor regression after intra-tumoral injection of LMWP-gelonin or TAT-gelonin conjugates. Tumor volumes were measured 30 days after cessation of drug treatment. Right: top to bottom represent tumors excised from mice treated with PBS solution (average tumor mass: 3.16±0.65 g); 100 μg gelonin (2.62±0.53 g); 110 μg LMWP-gelonin (0.33±0.12 g); 10 μg LMWP physically mixed with 100 μg gelonin (2.74±0.68 g); and 110 μg TAT-gelonin (0.21±0.04 g). (b) Tumor penetration of rhodamine-labeled free gelonin (top) or LWMP-gelonin conjugates (bottom) into the subcutaneously implanted tumor of a mouse. Ten hours after injection of the labeled drug or drug-conjugates, tumors were excised, processed into cryosections and examined by confocal microscopy. Left and right panels represent fluorescence microphotographs and DIC images, respectively (Reprinting with permission)
Fig.8  Effects of pH and silica content on the mucoadhesive properties of the nanocomposites. Mucoadhesion studies were performed via the rotating cylinder method in either HCl buffer (pH 2.0) or 100 mmol?L PBS buffer (pH 7.4) at 37°C. Data was presented as means±standard deviation of three experiments (Reprinting with permission)
Fig.8  Effects of pH and silica content on the mucoadhesive properties of the nanocomposites. Mucoadhesion studies were performed via the rotating cylinder method in either HCl buffer (pH 2.0) or 100 mmol?L PBS buffer (pH 7.4) at 37°C. Data was presented as means±standard deviation of three experiments (Reprinting with permission)
Fig.9  Effect of chitosan content on the release of amoxicillin from the silica-chitosan composites (Reprinting with permission)
Fig.9  Effect of chitosan content on the release of amoxicillin from the silica-chitosan composites (Reprinting with permission)
1 Capaldi B. Treatments and devices for future diabetes management. Nursing Times , 2005, 101(18): 30–32
2 Cobble M E. Initiating and intensifying insulin therapy for type 2 diabetes: why, when, and how. American Journal of Therapeutics , 2009, 16(1): 56–64
doi: 10.1097/MJT.0b013e3181966bf0
3 Prevention Cf DCa. National diabetes fact sheet general information and national estimates on diabetes in the United States. Centers for Disease Control and Prevention , 2003
4 Heinemann L. New ways of insulin delivery. International Journal of Clinical Practice. Supplement , 2011, 65(170): 31–46
doi: 10.1111/j.1742-1241.2010.02577.x
5 Gordon Still J. Development of oral insulin: progress and current status. Diabetes/Metabolism Research and Reviews , 2002, 18(S1): S29–S37
doi: 10.1002/dmrr.207
6 Heinemann L. New ways of insulin delivery. International Journal of Clinical Practice. Supplement , 2010, 64: 29–40
doi: 10.1111/j.1742-1241.2009.02276.x
7 Reis C P, Damge C. Nanotechnology as a promising strategy for alternative routes of insulin delivery. Methods in Enzymology , 2012, 508: 271–294
doi: 10.1016/B978-0-12-391860-4.00014-8
8 Fonte P, Andrade F, Araujo F, Andrade C, Neves J, Sarmento B. Chitosan-coated solid lipid nanoparticles for insulin delivery. Methods in Enzymology , 2012, 508: 295–314
doi: 10.1016/B978-0-12-391860-4.00015-X
9 Card J W, Magnuson B A. A review of the efficacy and safety of nanoparticle-based oral insulin delivery systems. American Journal of Physiology. Gastrointestinal and Liver Physiology , 2011, 301(6): G956–G967
doi: 10.1152/ajpgi.00107.2011
10 He P, Tang Z, Lin L, Deng M, Pang X, Zhuang X, Chen X. Novel biodegradable and pH-sensitive poly(ester amide) microspheres for oral insulin delivery. Macromolecular Bioscience , 2012, 12(4): 547–556
doi: 10.1002/mabi.201100358
11 Cefalu W T. Concept, strategies, and feasibility of noninvasive insulin delivery. Diabetes Care , 2004, 27(1): 239–246
doi: 10.2337/diacare.27.1.239
12 Krishnankutty R K, Mathew A, Sedimbi S K, Suryanarayan S, Sanjeevi C B. Alternative routes of insulin delivery. Zhong Nan Da Xue Xue Bao. Yi Xue Ban , 2009, 34(10): 933–948
13 Bellary S, Barnett A H. Inhaled insulin: new technology, new possibilities. International Journal of Clinical Practice , 2006, 60(6): 728–734
doi: 10.1111/j.1742-1241.2006.00976.x
14 Cefalu W T. Evolving strategies for insulin delivery and therapy. Drugs , 2004, 64(11): 1149–1161
doi: 10.2165/00003495-200464110-00001
15 Sajeesh S, Bouchemal K, Marsaud V, Vauthier C, Sharma C P. Cyclodextrin complexed insulin encapsulated hydrogel microparticles: An oral delivery system for insulin. Journal of Controlled Release , 2010, 147(3): 377–384
doi: 10.1016/j.jconrel.2010.08.007
16 Yadav N, Morris G, Harding S E, Ang S, Adams G G. Various non-injectable delivery systems for the treatment of diabetes mellitus. Endocrine, Metabolic & Immune Disorders Drug Targets , 2009, 9(1): 1–13
doi: 10.2174/187153009787582405
17 Banting F G, Best C H, Collip J B, Campbell W R, Fletcher A A. Pancreatic extracts in the treatment of diabetes mellitus. Canadian Medical Association Journal , 1922, 7: 6
18 Del Curto M D, Maroni A, Palugan L, Zema L, Gazzaniga A, Sangalli M E. Oral delivery system for two-pulse colonic release of protein drugs and protease inhibitor/absorption enhancer compounds. Journal of Pharmaceutical Sciences , 2011, 100(8): 3251–3259
doi: 10.1002/jps.22560
19 Jelvehgari M, Milani P Z, Siahi-Shadbad M R, Monajjemzadeh F, Nokhodchi A, Azari Z, Valizadeh H. In vitro and in vivo evaluation of insulin microspheres containing protease inhibitor. Arzneimittel-Forschung , 2011, 61(1): 14–22
doi: 10.1055/s-0031-1296163
20 Su F Y, Lin K J, Sonaje K, Wey S P, Yen T C, Ho Y C, Panda N, Chuang E Y, Maiti B, Sung H W. Protease inhibition and absorption enhancement by functional nanoparticles for effective oral insulin delivery. Biomaterials , 2012, 33(9): 2801–2811
doi: 10.1016/j.biomaterials.2011.12.038
21 Marschutz M K, Bernkop-Schnurch A. Oral peptide drug delivery: polymer-inhibitor conjugates protecting insulin from enzymatic degradation in vitro. Biomaterials , 2000, 21(14): 1499–1507
doi: 10.1016/S0142-9612(00)00039-9
22 Saudek C D. Novel forms of insulin delivery. Endocrinology and Metabolism Clinics of North America , 1997, 26(3): 599–610
doi: 10.1016/S0889-8529(05)70269-3
23 Avadi M R, Sadeghi A M, Mohammadpour N, Abedin S, Atyabi F, Dinarvand R, Rafiee-Tehrani M. Preparation and characterization of insulin nanoparticles using chitosan and Arabic gum with ionic gelation method. Nanomedicine; Nanotechnology, Biology, and Medicine , 2010, 6(1): 58–63
doi: 10.1016/j.nano.2009.04.007
24 Cui F, He C, He M, Tang C, Yin L, Qian F, Yin C. Preparation and evaluation of chitosan-ethylenediaminetetraacetic acid hydrogel films for the mucoadhesive transbuccal delivery of insulin. Journal of Biomedical Materials Research. Part A , 2009, 89A(4): 1063–1071
doi: 10.1002/jbm.a.32071
25 Cui F, Qian F, Zhao Z, Yin L, Tang C, Yin C. Preparation, characterization, and oral delivery of insulin loaded carboxylated chitosan grafted poly(methyl methacrylate) nanoparticles. Biomacromolecules , 2009, 10(5): 1253–1258
doi: 10.1021/bm900035u
26 Schilling R, Mitra A. Degradation of insulin by trypsin and alpha-chymotrypsin. Pharmaceutical Research , 1991, 8(6): 721–727
doi: 10.1023/A:1015893832222
27 Nishihata T, Rytting J H, Kamada A, Higuchi T. Enhanced intestinal absorption of insulin in rats in the presence of sodium 5-methoxysalicylate. Diabetes , 1981, 30(12): 1065–1067
doi: 10.2337/diabetes.30.12.1065
28 Cui C Y, Lu W L, Xiao L, Zhang S Q, Huang Y B, Li S L, Zhang R J, Wang G L, Zhang X, Zhang Q. Sublingual delivery of insulin: effects of enhancers on the mucosal lipid fluidity and protein conformation, transport, and in vivo hypoglycemic activity. Biological & Pharmaceutical Bulletin , 2005, 28(12): 2279–2288
doi: 10.1248/bpb.28.2279
29 Muranishi S. Delivery system design for improvement of intestinal absorption of peptide drugs. Yakugaku Zasshi , 1997, 117(7): 394–414
30 Chung S W, Hil-lal T A, Byun Y. Strategies for non-invasive delivery of biologics. Journal of Drug Targeting , 2012, 20(6): 481–501
doi: 10.3109/1061186X.2012.693499
31 Schwarze S R, Ho A, Vocero-Akbani A, Dowdy S F. In vivo protein transduction: delivery of a biologically active protein into the mouse. Science , 1999, 285(5433): 1569–1572
doi: 10.1126/science.285.5433.1569
32 Schwarze S R, Dowdy S F. In vivo protein transduction: intracellular delivery of biologically active proteins, compounds and DNA. Trends in Pharmacological Sciences , 2000, 21(2): 45–48
doi: 10.1016/S0165-6147(99)01429-7
33 Cooper I, Sasson K, Teichberg V I, Schnaider-Beeri M, Fridkin M, Shechter Y. Peptide derived from HIV-1 TAT protein, destabilizes a monolayer of endothelial cells in an in vitro model of the blood-brain barrier, and allows permeation of high molecular weight proteins. Journal of Biological Chemistry , 2012, ???:
doi: 10.1074/jbc.M112.395384
34 Yu R, Zeng Z, Guo X, Zhang H, Liu X, Ding Y, Chen J. The TAT peptide endows PACAP with an enhanced ability to traverse bio-barriers. Neuroscience Letters , 2012, 527(1): 1–5
doi: 10.1016/j.neulet.2012.08.005
35 Elliott G, O'Hare P. Intercellular trafficking and protein delivery by a herpesvirus structural protein. Cell , 1997, 88(2): 223–233
doi: 10.1016/S0092-8674(00)81843-7
36 Min S H, Kim D M, Kim M N, Ge J, Lee D C, Park I Y, Park K C, Hwang J S, Cho C W, Yeom Y I. Gene delivery using a derivative of the protein transduction domain peptide, K-Antp. Biomaterials , 2010, 31(7): 1858–1864
doi: 10.1016/j.biomaterials.2009.11.019
37 Derossi D, Joliot A H, Chassaing G, Prochiantz A. The third helix of the antennapedia homeodomain translocates through biological membranes. Journal of Biological Chemistry , 1994, 269(14): 10444–10450
38 Jin G S, Zhu G D, Zhao Z G, Liu F S. VP22 enhances the expression of glucocerebrosidase in human Gaucher II fibroblast cells mediated by lentiviral vectors. Clinical and Experimental Medicine , 2012, 12(3): 135–143
doi: 10.1007/s10238-011-0152-7
39 Tanaka M, Kato A, Satoh Y, Ide T, Sagou K, Kimura K, Hasegawa H, Kawaguchi Y. Herpes simplex virus 1 VP22 regulates translocation of multiple viral and cellular proteins and promotes neurovirulence. Journal of Virology , 2012, 86(9): 5264–5277
doi: 10.1128/JVI.06913-11
40 Chang L C, Lee H F, Yang Z, Yang V. Low molecular weight protamine (LMWP) as nontoxic heparin/low molecular weight heparin antidote (I): Preparation and characterization. AAPS PharmSci , 2001, 3(3): 7–14
doi: 10.1208/ps030317
41 Chang L C, Liang J, Lee H F, Lee L, Yang V. Low molecular weight protamine (LMWP) as nontoxic heparin/low molecular weight heparin antidote (II): In vitro evaluation of efficacy and toxicity. AAPS PharmSci , 2001, 3(3): 15–23
doi: 10.1208/ps030318
42 Chang L C, Wrobleski S, Wakefield T, Lee L, Yang V. Low molecular weight protamine as nontoxic heparin/low molecular weight heparin antidote (III): Preliminary in vivo evaluation of efficacy and toxicity using a canine model. AAPS PharmSci , 2001, 3(3): 24–31
doi: 10.1208/ps030319
43 Park Y J, Chang L C, Liang J F, Moon C, Chung C P, Yang V C. Nontoxic membrane translocation peptide from protamine, low molecular weight protamine (LMWP), for enhanced intracellular protein delivery: in vitro and in vivo study. FASEB Journal , 2005, 19(11): 1555–1557
44 Xia H, Gao X, Gu G, Liu Z, Zeng N, Hu Q, Song Q, Yao L, Pang Z, Jiang X, Chen J, Chen H. Low molecular weight protamine-functionalized nanoparticles for drug delivery to the brain after intranasal administration. Biomaterials , 2011, 32(36): 9888–9898
doi: 10.1016/j.biomaterials.2011.09.004
45 Ramadas M W P, Dileep K J, Ramadas M, Anitha Y, Sharma C P, 0. Lipoinsulin encapsulated alginate-chitosan capsules: intestinal delivery in diabetic rats. Journal of Microencapsulation , 2000, 17(4): 405–411
doi: 10.1080/026520400405660
46 Kimura T, Sato K, Sugimoto K, Tao R, Murakami T, Kurosaki Y, Nakayama T. Oral administration of insulin as poly(vinyl alcohol)-gel spheres in diabetic rats. Biological & Pharmaceutical Bulletin , 1996, 19(6): 897–900
doi: 10.1248/bpb.19.897
47 Mitchell D J, Steinman L, Kim D T, Fathman C G, Rothbard J B. Polyarginine enters cells more efficiently than other polycationic homopolymers. Journal of Peptide Research , 2000, 56(5): 318–325
doi: 10.1034/j.1399-3011.2000.00723.x
48 Futaki S, Nakase I, Suzuki T, Zhang, Sugiura Y. Translocation of branched-chain arginine peptides through cell membranes: flexibility in the spatial disposition of positive charges in membrane-permeable peptides. Biochemistry , 2002, 41(25): 7925–7930
doi: 10.1021/bi0256173
49 Wong T W. Chitosan and its use in design of insulin delivery system. Recent Patents on Drug Delivery & Formulation , 2009, 3(1): 8–25
doi: 10.2174/187221109787158346
50 Damge C, Maincent P, Ubrich N. Oral delivery of insulin associated to polymeric nanoparticles in diabetic rats. Journal of Controlled Release , 2007, 117(2): 163–170
doi: 10.1016/j.jconrel.2006.10.023
51 Sarmento B, Ribeiro A, Veiga F, Sampaio P, Neufeld R, Ferreira D. Alginate/chitosan nanoparticles are effective for oral insulin delivery. Pharmaceutical Research , 2007, 24(12): 2198–2206
doi: 10.1007/s11095-007-9367-4
52 Liang J F, Zhen L, Chang L C, Yang V C. A less toxic heparin antagonist—low molecular weight protamine. Biochemistry. Biokhimiia , 2003, 68(1): 116–120
doi: 10.1023/A:1022109905487
53 Tsui B, Singh V K, Liang J F, Yang V C. Reduced reactivity towards anti-protamine antibodies of a low molecular weight protamine analogue. Thrombosis Research , 2001, 101(5): 417–420
doi: 10.1016/S0049-3848(00)00427-8
54 Carlsson J, Drevin H, Axén R. Protein thiolation and reversible protein-protein conjugation.N-Succinimidyl 3-(2-pyridyldithio)propionate, a new heterobifunctional reagent. Biochemical Journal , 1978, 173(3): 723–737
55 Chickering D E, Mathiowitz E. Bioadhesive microspheres I. A novel electrobalance-based method to study adhesive interactions between individual microspheres and intestinal mucosa. Journal of Controlled Release , 1995, 34(3): 251–262
doi: 10.1016/0168-3659(95)00011-V
56 Sudhakar Y, Kuotsu K, Bandyopadhyay A K. Buccal bioadhesive drug delivery—a promising option for orally less efficient drugs. Journal of Controlled Release , 2006, 114(1): 15–40
doi: 10.1016/j.jconrel.2006.04.012
[1] Jennica L. Zaro,Wei-Chiang Shen. Cationic and amphipathic cell-penetrating peptides (CPPs): Their structures and in vivo studies in drug delivery[J]. Front. Chem. Sci. Eng., 2015, 9(4): 407-427.
[2] Huining HE, Qiuling LIANG, Meong Cheol SHIN, Kyuri LEE, Junbo GONG, Junxiao YE, Quan LIU, Jingkang WANG, Victor YANG. Significance and strategies in developing delivery systems for bio-macromolecular drugs[J]. Front Chem Sci Eng, 2013, 7(4): 496-507.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed