Editorial Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jun 21, 2012; 18(23): 2895-2901
Published online Jun 21, 2012. doi: 10.3748/wjg.v18.i23.2895
Potential prospects of nanomedicine for targeted therapeutics in inflammatory bowel diseases
Madharasi VA Pichai, Lynnette R Ferguson, Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
Author contributions: All authors have contributed equally to this paper.
Correspondence to: Lynnette R Ferguson, Professor, Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton Private Bag 92019, Auckland Mail Centre, Auckland 1142, New Zealand. l.ferguson@auckland.ac.nz
Telephone: +64-9-9236372 Fax: +64-9-3035962
Received: October 29, 2011
Revised: April 5, 2012
Accepted: April 10, 2012
Published online: June 21, 2012

Abstract

Inflammatory bowel diseases (IBDs) such as Crohn’s disease are highly debilitating. There are inconsistencies in response to and side effects in the current conventional medications, failures in adequate drug delivery, and the lack of therapeutics to offer complete remission in the presently available treatments of IBD. This suggests the need to explore beyond the horizons of conventional approaches in IBD therapeutics. This review examines the arena of the evolving IBD nanomedicine, studied so far in animal and in vitro models, before comprehensive clinical testing in humans. The investigations carried out so far in IBD models have provided substantial evidence of the nanotherapeutic approach as having the potential to overcome some of the current drawbacks to conventional IBD therapy. We analyze the pros and cons of nanotechnology in IBD therapies studied in different models, aimed at different targets and mechanisms of IBD pathogenesis, in an attempt to predict its possible impact in humans.

Key Words: Inflammatory bowel disease, Crohn’s disease, Ulcerative colitis, Tumor necrosis factor-α, Nanomedicine



INTRODUCTION

Crohn’s disease (CD) and ulcerative colitis (UC) constitute the two principal components of inflammatory bowel diseases (IBDs), which occur as a result of dysregulated immune responses in genetically predisposed individuals due to various environmental conditions[1]. There are sufficient similarities in the pathological conditions in CD and UC that cause about 10% of IBD cases to be diagnosed as indeterminate IBD[2]. Nevertheless, CD and UC show discrete risk factors and dissimilar gene and protein expressions, which manifest distinctive pathophysiological mechanisms. CD exhibits a transmural inflammatory response and can be associated with granulomas, whereas UC usually shows mucosa-confined inflammation[2-8]. Genomic technologies are now being used to separate the effects of different susceptibility genes in the two diseases. For example, Wu et al[6] have studied 36 expression profiles of colonoscopic pinch biopsies from CD and UC patients. Affected genes, mostly related to interferon (IFN)-γ inducible T helper cell 1 (TH1) process and antigen presentation in CD patients, were differentially regulated, with the upregulation of 47 genes and downregulation of 30 genes. In contrast, the expression of genes from UC biopsies showed upregulation of 51 genes and downregulation of 81 other genes, associated with biosynthesis, metabolism and electrolyte transport[6].

The common conventional medications currently in use to treat both CD and UC involve 5-aminosalicylic acid drugs, corticosteroids, immunosuppressive agents, biologic therapies and antibiotics[9], with a customary “step up” approach of starting with aminosalicylates and rising to corticosteroids and immunosuppressive agents in response to the persisting conditions of the disease. The more effective biological therapies are usually considered as a last option and only in case of refractory diseases, because their systemic action in the host often leads to adverse effects[10,11].

Nanomedicines are precise therapeutics established with the aid of nanotechnology to treat diseases at the molecular level[12]. The application of nanotechnology in medicine can be termed as nanomedicine. It is an evolving face of medicine that uses nanoparticulate carriers to deliver therapeutics targeted to specific cells, or constituents of cells or tissues. Studies have shown nanomedicines to be more beneficial than conventional medications, because their size leads to more effecting targeting, better availability at diseased tissues, and decreased adverse effects. Moreover, nanomedicines have been found to have similar or even better therapeutic impacts at lower drug concentrations than their conventional counterparts[12]. However, although the arena of nanomedicine appears to be encouraging for IBD therapy, concerns related to the impact of the nature of nanoparticles due to their size, shape, aggregation potential, and surface chemistry on the IBD gut need to be scrutinized[12,13], and investigations on the impact of nanomedicine in IBD therapy is currently in early stages. As targeted drug or biological delivery to sites of inflammation remains a crucial challenge in the current treatment of IBD[14], nanostrategies involving short interfering RNAs (siRNAs), antisense oligonucleotides, nanomedicines delivered to the sites of malfunction in IBD can be a valuable therapeutic approach.

The RNA interference technique, notable for specificity, can be speculated to regulate the expression of proinflammatory cytokines and genes related to IBD at the mRNA level[15]. As the impact of noncoding RNAs and RNA silencing in gene modulation is known to be great[16], the use of siRNAs as drugs to silence proinflammatory genes is being scrutinized in various animal models of IBD. This strategy also reduces the chances of immune reactions usually associated with viral vectors[15,17,18]. Due to the potential importance of targeted therapy in IBD, this review is presented to explore the advancements in the prospects of nanomedicine in the modulation of gene expression and targeted therapeutics in IBD (Figure 1).

Figure 1
Figure 1 Graphical representation of nanoinvestigations in inflammatory bowel diseases models. IBD: Inflammatory bowel disease.
GENE AND PROTEIN MODULATIONS WITH NANO PROSPECTS

Amongst the key genes involved in IBD pathogenesis, the function of tumor necrosis factor (TNF)-α in the mediation of inflammation in IBD is extensively acknowledged. Therefore, many biological therapies comprising monoclonal antibodies or soluble receptors are intended to reduce TNF-α activity, and have been extensively tested in many clinical trials[19-24]. However, there are adverse side effects due to the systemic depletion of TNF-α. These adverse effects involve amplified infusion reactions, immunosuppression, opportunistic infections and decreased efficacy of the biologics due to antibody formation against them[24-26].

The gene silencing nanostrategy, in which orally delivered TNF-α siRNA is encapsulated in thioketal nanoparticles (TKNs) made from the polymer poly-PPADT (1, 4-phenyleneacetone dimethylene thioketal), effectively decreases the levels of TNF-α mRNA levels at sites of intestinal inflammation in dextran sulfate sodium (DSS)-induced mouse models of UC. In this study, the site specific delivery of siRNA was made possible due to the ability of TKNs to degrade in the presence of higher levels of reactive oxygen species (ROS) present in regions of inflammation in the intestinal tissue[27]. In another study, TNF-α siRNA/polyethyleneimine (PEI) nanocomplex was shown to inhibit TNF-α secretion by macrophages in vitro, whereas the oral administration of TNF-α siRNA/PEI nanocomplexes in lipopolysaccharides (LPS)-treated mice models was found to reduce specifically the synthesis and secretion of TNF-α in the colon[28]. Nanoparticles in a microsphere oral system (NiMOS), comprised of TNF-α siRNA entrapped in type B gelatin enclosed in poly(e-caprolactone) (PCL) microspheres, were found to exhibit favorable gene silencing in the colon tissues of DSS-treated murine models of UC. This treatment results in the suppression of proinflammatory cytokines such as interleukin (IL)-1β, IFN-γ, chemokine monocyte chemoattractant protein (MCP)-1, permitting an increase in body weight and diminished action of tissue myeloperoxidase in mouse models[29]. A protein modulation nanostrategy involving monovalent and bivalent murine TNF-α neutralizing nanobody proteins has been investigated in DSS-induced murine chronic colitis models. Lactococcus lactis engineered to produce the therapeutic nanobodies was orally administered, which resulted in a significant reduction in the TNF-α driven inflammation in the mucosa of the colon in mouse models, without affecting considerable TNF-α levels in the systemic circulation[30].

Increased TNF-α suppresses the expression of the anti-inflammatory protein prohibitin (PHB) in IBD[31,32], therefore, a study by Theiss et al[33] considered the oral delivery of PHB entrapped in poly (lactic acid) nanoparticles in mouse models of DSS-induced colitis. This strategy inhibited the TNF-α-induced nuclear factor (NF)-κB activation; consequently curtailing inflammatory reactions and reducing the severity of colitis. Double-stranded decoy oligonucleotides (ODNs) against the proinflammatory NF-κB gene were enclosed in chitosan-modified poly (D,L-lactide-co-glycolide) nanospheres (CS-PLGA NSs) and delivered orally to DSS-induced murine colitis models. This study showed the absorption of the ODN- CS-PLGA NSs in inflamed mucosal regions, producing considerable curative effects on DSS-induced diarrhea, bloody feces, shortening of colon length, and myeloperoxidase activity[34].

Besides directly inhibiting the TNF-α gene in macrophages, macrophages more generally play a role in inducing the pathogenic inflammatory reactions[35]. This study has revealed the importance of mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4) gene in macrophages in mediating the production of inflammatory cytokines. Map4k4 siRNA encapsulated in β1,3-D-glucan shells silenced Map4k4 expression in vivo in mice treated with LPS, protecting them from LPS-induced systemic inflammation by suppressing the production of TNF-α and IL-1β[35].

Matrix metalloproteinases (MMPs) play a vital role in tissue remodeling by regulating the intestinal tissue architecture during the inflammatory reactions and wound healing in IBD[36,37]. Studies have indicated the increased expression of MMP-3 (stromelysin-1) and MMP-10 (stromelysin-2) in causing enhanced tissue injury in DSS-induced murine colitis[38,39]. Furthermore, IBD patients have shown increased MMP-3 and MMP-10 expression in the gut and intestinal ulcer tissues[39-42]. Polymorphisms in various MMP genes may be susceptibility factors for IBD risk, at least in some populations[43]. A study by Kobayashi et al[39]

demonstrated the specific inhibition of MMP-3 and MMP-10 by siRNA targeted against MMP-3 and MMP-10, having a therapeutic benefit in protecting the colon tissue and reducing the severity of colitis in DSS-treated murine models, which could therefore be a valuable gene silencing option to prevent intestinal damage in IBD (Figure 2).

Figure 2
Figure 2 Nanomodulations whose efficacy has been validated in animal models of inflammatory bowel diseases. Genes regulated therapeutically by nano gene silencing in intestinal tissues and macrophages and protein nanobodies that have been investigated to have therapeutic impacts to help control inflammation and tissue destruction in animal models relevant to inflammatory bowel diseases (IBDs). TNF: Tumor necrosis factor; Map4k4: Mitogen-activated protein kinase kinase kinase kinase 4; MMP: Matrix metalloproteinase; NF-κB: Nuclear factor kappa B.

Cyclin D1 (CyD1) is a cell cycle regulatory protein that is upregulated in IBD in both epithelial and immune cells[44]. A leukocyte-directed siRNA against CyD1 mRNA inhibits the intestinal inflammatory responses in murine models of DSS-induced colitis. Silencing the CyD1 gene decreases the induction of TH1 cell inflammatory cytokines TNF-α and IL-12, but has no impact on the production of TH2 cell cytokine IL-10[45]. Therapeutic efforts to enhance the action of the anti-inflammatory cytokine IL-10, which is known to be critically involved in maintaining mucosal immune balance due to its potent impact on immunosuppression[46] and involvement in CD pathogenesis[47,48], have been largely unsuccessful to date. This is thought to be due to the adverse side effects caused by systemic action of the IL-10 therapies, and the low concentrations of IL-10 delivered to the intestinal tissues[49]. Therefore, biologics intending to enhance cytokine IL-10 action have been dropped from the current IBD therapies[50]. However, because the involvement of IL-10 and its genetic variations in IBD is great[47,48,51], a consideration of the targeted study by Bhavsar et al[52], which involved the nanodelivery of IL-10-producing plasmid to the mucosa in murine models mimicking IBD intestinal epithelial pathogenesis[53] can be scrutinized. According to this study, trinitrobenzene sulfonic acid (TNBS)-induced acute colitis models in Balb/c mice were treated with NiMOS intended for oral gene therapy. This comprised the pORF5-mIL-10 plasmid DNA encapsulated in type B gelatin nanoparticles in PCL. This strategy directed the local transfection of IL-10 plasmid in inflamed intestinal tissues and caused its enhanced expression, which led to suppression of predominant proinflammatory cytokines such as IFN-γ, TNF-α, IL-1α, IL-1β and IL-12, consequently causing the therapeutic benefits of restored colon length and weight, increased body weight, and beneficial clinical activity score[52].

IMPLICATIONS OF IBD DRUGS GOING NANO

Nanomedicines comprising IBD drugs loaded onto na-noparticles, designed to cope and act in accordance with the pathophysiological changes in the intestinal tissues of IBD, can be an intelligent mode of targeted drug delivery. This approach offers the possibility of eliminating undesirable side effects usually caused by systemic action of the drugs[14]. The usual pathophysiological conditions related to inflamed intestinal tissues in IBD predominantly involve abnormal intestinal permeability, increased presence of immune cells, and higher levels of mucus production[54-56].

Cellular interaction of nanoparticles in the IBD gut

Nanomedicines in IBD can potentially be more efficient in their mechanism due to the cellular intake of the nanoparticles by the cells at the targeted sites of delivery. This means that they are not eliminated from the intestinal tract by diarrhea, as are many current conventional medications. This is an important IBD symptom[57,58]. Nanoparticles in the gastrointestinal tract are usually found to be adsorbed either by paracellular transport or endocytosis by regular epithelial cells[59]. Specialized differentiated epithelial cells called M cells, which form major populations of Peyer’s patches are involved in the predominant uptake of nanoparticles through transcytosis[60,61]. Predominant CD mutations such as R702W, G908R and 3020insC have been associated with ileal-specific disease[62,63], which show an enhanced presence in Peyer’s patches and M cells, which may cause an increase in the uptake of dietary and nanoparticulate substances[13]. In addition to these, translocation of nanoparticles in the intestinal tract can also occur by persorption through gaps or holes at the villous tips[64,65]. Cells involve the autophagic mechanism to cause the clearance of nanoparticles[66], and Powell et al[13] have indicated that mutations in the autophagy gene Atg16L1 in IBD subjects can be susceptible to possible alterations in the clearance of nanoparticles.

Investigations of IBD drugs in nanomodes

Furthermore, IBD drugs delivered in nanomodes have been shown to have greater therapeutic impacts as compared to their conventional delivery studied in animal models, For example, the anti-inflammatory IBD drug mesalamine (5-ASA) covalently linked to the PCL nanoparticles was found to be 60 times more efficient as a nanomedicine at much lower doses (0.5 mg/kg) than the free solution of 5-ASA (30 mg/kg) in treating TNBS-induced colitis in BALB/c murine models[67]. Moulari et al[68] have established that silicon nanoparticles have a sixfold increased ability to adhere to inflamed tissues when compared to tissues in healthy controls. In this study, 5-ASA loaded in its methylated form in silicon nanoparticles was shown to collect in inflamed regions in TNBS-induced murine colitis models, inducing a positive impact on clinical activity score and myeloperoxidase activity at reduced drug doses, as compared to conventional delivery[68]. An immunosuppressive drug tacrolimus, used to treat UC, was encapsulated in polylactic-co-glycolic acid (PLGA) nanoparticles and used to treat murine models of TNBS- and oxazolone-induced colitis. This study showed that nanomedicine had an augmented and specific action with a threefold increased penetration in inflamed tissues when compared to healthy tissues[69]. Also, tacrolimus-loaded PLGA nanoparticles and tacrolimus-loaded pH-sensitive Eudragit P-4135F nanoparticles showed diminished side effects in DSS-induced murine colitis models when compared to the free tacrolimus which causes nephrotoxicity in traditional delivery[70]. An anti-inflammatory tripeptide Lys-Pro-Val (KPV) loaded into polylactide (PLA) nanoparticles delivered in combination with a polysaccharide hydrogel had a similar anti-inflammatory effect at 12 000-fold lower doses (25.2 ng/d) to that of KPV in free solution (200 μg/d), thus demonstrating the greater therapeutic efficiency of the nanomode of the drug in treating DSS-induced colitis in murine models[71]. Nakase et al[72] demonstrated that dexamethasone, encapsulated in poly-DL-lactic acid (PDLLA) microspheres was more effective in ameliorating DSS-induced murine colitis when compared to the free solution of the same drug, because the microsphere form was engulfed by the immune cells in the inflamed colonic tissue, which resulted in increased efficiency of the drug in mouse models. An ex vivo study by Serpe et al[73] showed solid lipid nanoparticles (SLNs) comprising the anti-inflammatory molecule cholesteryl butyrate (chol-but) showed a greater impact than butyrate alone in significantly reducing proinflammatory cytokines such as IL-1β and TNF-α and increasing IL-10 production in whole blood ex vivo models of peripheral blood mononuclear cells (PBMCs) obtained from IBD patients taking no anti-inflammatory medications. Furthermore, this study demonstrated SLNs, consisting of the immunosuppressive corticosteroid dexamethasone, suppressed TNF-α by 90% when the free solution of dexamethasone showed a TNF-α suppression of 25% at the highest concentrations in similar whole blood IBD ex vivo models. These studies provide preliminary support for the effects of SLNs chol-but and SLN dexamethasone in inducing an enhanced anti-inflammatory activity, due to the more effective cellular intake of the nanodrug forms, as compared to the free drugs in solution (Table 1).

Table 1 Comparison of therapeutic parameters.
Experimental systemDrug in nano modeComparison of differences in certain distinct therapeutic parameters
Ref.
Nano modeControls
In vivo TNBS-induced murine colitis5-ASA covalently linked to PCL nanoparticlesMyeloperoxidase (MPO) activity of 5-ASA-NP at 0.5 mg/kg: 15.2 ± 5.6 U/mgMPO activity of 5-ASA free solution at 30 mg/kg: 16.2 ± 3.6 U/mg[67]
In vivo TNBS-induced murine colitis5-ASA in silicon nanoparticlesMPO activity of 5-ASA-Si NP at 25 mg/kg: 5.2 ± 2.4 U/mgMPO activity of 5-ASA-free solution at 100 mg/kg: 8.2 ± 3.4 U/mg[68]
In vivo TNBS-induced murine colitis/oxazolone-induced murine colitisTacrolimus in PLGA NPsEnhanced penetration into the inflamed tissue-FK506-NP, 105 ± 24 nmol/cm2Healthy tissue penetration-FK506-NP, 51 ± 13 nmol/cm2[69]
In vivo DSS- induced murine colitisTacrolimus in PLGA/or pH sensitive Eudragit P-4135F NPsDiminished side effectsIncreased susceptibility to nephrotoxicity[70]
In vivo DSS-induced murine colitisAnti-inflammatory tripeptide KPV in PLA NPsNanomode with lowered doses at 25.2 ng/dFree solution has the similar anti-inflammatory impact at 200 μg/d[71]
Whole blood ex vivo models of PBMCsDexamethasone in SLNs90% reduction in proinflammatory cytokines IL-1β and TNF-α25% reduction in TNF-α by the free solution[73]
Whole blood ex vivo models of PBMCschol-but in SLNsSignificant decrease in IL-1β, and TNF-α increase in IL-10-[73]
LIKELY CONCERNS OF GOING NANO

The general concern associated with the nano approach is due to the fact that nano-sized materials display altered physicochemical properties[74] as compared to their larger counterparts, with chances of causing possible toxicity, since nonbiological nanoparticulate carriers above a particle size of 100-200 nm can alter normal cellular activity, because they can invoke cell membrane ruffling, cytoskeletal rearrangement and stimulate endocytic machinery causing their ingression in phagocytic cells[75]. However, reliable data on the adverse impacts of nanomedicine in IBD is unavailable, whereas the impact of nanoparticles themselves in the gastrointestinal tract might vary according to the nanoparticle polymer material and nanoparticle size, as surface interactions and surface chemistry differ for different nanoparticle sizes. Also, different nanoparticle sizes can cause different mechanisms of cellular uptake, due to which, nanoparticle sizes can be modulated to cause different intracellular effects[13,76,77]. The studies on the effects of nanoparticles themselves in the human gastrointestinal tract in IBD have been limited and need to be explored further.

Although nanostrategies for IBD therapeutics investigated in animal and in vitro models of IBD have shown promise, it is still only the dawn of the era of interest in IBD nanomedicine, and there is a definite need for further extensive investigations on many issues related to the safety and uptake of the different nanomedical therapeutics acting on various pathways and phases in the human gastrointestinal tract. It is essential to be confident of their consequent impact on immune responses and therapeutic effects in the different genotypic populations, before recommending the clinical use of nanomedicines to treat IBD in humans.

Footnotes

Peer reviewers: Dr. Pingchang Yang, MD, PhD, Department of Pathology and Molecular Medicine, McMaster University, BBI-T3330, 50 Charlton Ave East, Hamilton, L8N 4A6, Canada; Emiko Mizoguchi, MD, PhD, Department of Medicine, Gastrointestinal Unit, GRJ 702, Massachusetts General Hospital, Boston, MA 02114, United States

S- Editor Cheng JX L- Editor Kerr C E- Editor Zheng XM

References
1.  Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448:427-434.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Podolsky DK. Inflammatory bowel disease. N Engl J Med. 2002;347:417-429.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, Brant SR, Silverberg MS, Taylor KD, Barmada MM. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet. 2008;40:955-962.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Parkes M, Jewell D. Ulcerative colitis and Crohns disease: molecular genetics and clinical implications. Expert Rev Mol Med. 2001;2001:1-18.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  McGovern DP, Gardet A, Törkvist L, Goyette P, Essers J, Taylor KD, Neale BM, Ong RT, Lagacé C, Li C. Genome-wide association identifies multiple ulcerative colitis susceptibility loci. Nat Genet. 2010;42:332-337.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Wu F, Dassopoulos T, Cope L, Maitra A, Brant SR, Harris ML, Bayless TM, Parmigiani G, Chakravarti S. Genome-wide gene expression differences in Crohn's disease and ulcerative colitis from endoscopic pinch biopsies: insights into distinctive pathogenesis. Inflamm Bowel Dis. 2007;13:807-821.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Shkoda A, Werner T, Daniel H, Gunckel M, Rogler G, Haller D. Differential protein expression profile in the intestinal epithelium from patients with inflammatory bowel disease. J Proteome Res. 2007;6:1114-1125.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Lawrance IC, Fiocchi C, Chakravarti S. Ulcerative colitis and Crohn's disease: distinctive gene expression profiles and novel susceptibility candidate genes. Hum Mol Genet. 2001;10:445-456.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Talley NJ, Abreu MT, Achkar JP, Bernstein CN, Dubinsky MC, Hanauer SB, Kane SV, Sandborn WJ, Ullman TA, Moayyedi P. An evidence-based systematic review on medical therapies for inflammatory bowel disease. Am J Gastroenterol. 2011;106 Suppl 1:S2-25; quiz S26.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Desilva S, Kaplan G, Panaccione R. Sequential therapies for Crohn's disease: optimizing conventional and biologic strategies. Rev Gastroenterol Disord. 2008;8:109-116.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Schmidt KJ, Büning J, Jankowiak C, Lehnert H, Fellermann K. Crohn's targeted therapy: myth or real goal? Curr Drug Discov Technol. 2009;6:290-298.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Hock SC, Ying YM, Wah CL. A review of the current scientific and regulatory status of nanomedicines and the challenges ahead. PDA J Pharm Sci Technol. 2011;65:177-195.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Powell JJ, Faria N, Thomas-McKay E, Pele LC. Origin and fate of dietary nanoparticles and microparticles in the gastrointestinal tract. J Autoimmun. 2010;34:J226-J233.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Lamprecht A. IBD: selective nanoparticle adhesion can enhance colitis therapy. Nat Rev Gastroenterol Hepatol. 2010;7:311-312.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Plevy SE, Targan SR. Future therapeutic approaches for inflammatory bowel diseases. Gastroenterology. 2011;140:1838-1846.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Ferguson LR. RNA silencing: Mechanism, biology and responses to environmental stress. Mutat Res. 2011;714:93-94.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Pellish RS, Nasir A, Ramratnam B, Moss SF. Review article: RNA interference--potential therapeutic applications for the gastroenterologist. Aliment Pharmacol Ther. 2008;27:715-723.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Lieberman J, Song E, Lee SK, Shankar P. Interfering with disease: opportunities and roadblocks to harnessing RNA interference. Trends Mol Med. 2003;9:397-403.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Mueller C. Tumour necrosis factor in mouse models of chronic intestinal inflammation. Immunology. 2002;105:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Holtmann MH, Neurath MF. Anti-TNF strategies in stenosing and fistulizing Crohn's disease. Int J Colorectal Dis. 2005;20:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  D'Haens G. Anti-TNF therapy for Crohn's disease. Curr Pharm Des. 2003;9:289-294.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Oldenburg B, Hommes D. Biological therapies in inflammatory bowel disease: top-down or bottom-up? Curr Opin Gastroenterol. 2007;23:395-399.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Sandborn WJ. Strategies for targeting tumour necrosis factor in IBD. Best Pract Res Clin Gastroenterol. 2003;17:105-117.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  van Deventer SJ. New biological therapies in inflammatory bowel disease. Best Pract Res Clin Gastroenterol. 2003;17:119-130.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Van Assche G, Vermeire S, Rutgeerts P. Safety issues with biological therapies for inflammatory bowel disease. Curr Opin Gastroenterol. 2006;22:370-376.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Hoentjen F, van Bodegraven AA. Safety of anti-tumor necrosis factor therapy in inflammatory bowel disease. World J Gastroenterol. 2009;15:2067-2073.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Wilson DS, Dalmasso G, Wang L, Sitaraman SV, Merlin D, Murthy N. Orally delivered thioketal nanoparticles loaded with TNF-α-siRNA target inflammation and inhibit gene expression in the intestines. Nat Mater. 2010;9:923-928.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Laroui H, Theiss AL, Yan Y, Dalmasso G, Nguyen HT, Sitaraman SV, Merlin D. Functional TNFα gene silencing mediated by polyethyleneimine/TNFα siRNA nanocomplexes in inflamed colon. Biomaterials. 2011;32:1218-1228.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kriegel C, Amiji M. Oral TNF-α gene silencing using a polymeric microsphere-based delivery system for the treatment of inflammatory bowel disease. J Control Release. 2011;150:77-86.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Vandenbroucke K, de Haard H, Beirnaert E, Dreier T, Lauwereys M, Huyck L, Van Huysse J, Demetter P, Steidler L, Remaut E. Orally administered L. lactis secreting an anti-TNF Nanobody demonstrate efficacy in chronic colitis. Mucosal Immunol. 2010;3:49-56.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Theiss AL, Idell RD, Srinivasan S, Klapproth JM, Jones DP, Merlin D, Sitaraman SV. Prohibitin protects against oxidative stress in intestinal epithelial cells. FASEB J. 2007;21:197-206.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Hsieh SY, Shih TC, Yeh CY, Lin CJ, Chou YY, Lee YS. Comparative proteomic studies on the pathogenesis of human ulcerative colitis. Proteomics. 2006;6:5322-5331.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Theiss AL, Laroui H, Obertone TS, Chowdhury I, Thompson WE, Merlin D, Sitaraman SV. Nanoparticle-based therapeutic delivery of prohibitin to the colonic epithelial cells ameliorates acute murine colitis. Inflamm Bowel Dis. 2011;17:1163-1176.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Tahara K, Samura S, Tsuji K, Yamamoto H, Tsukada Y, Bando Y, Tsujimoto H, Morishita R, Kawashima Y. Oral nuclear factor-κB decoy oligonucleotides delivery system with chitosan modified poly(D,L-lactide-co-glycolide) nanospheres for inflammatory bowel disease. Biomaterials. 2011;32:870-878.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature. 2009;458:1180-1184.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Naito Y, Yoshikawa T. Role of matrix metalloproteinases in inflammatory bowel disease. Mol Aspects Med. 2005;26:379-390.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Ravi A, Garg P, Sitaraman SV. Matrix metalloproteinases in inflammatory bowel disease: boon or a bane? Inflamm Bowel Dis. 2007;13:97-107.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  von Lampe B, Barthel B, Coupland SE, Riecken EO, Rosewicz S. Differential expression of matrix metalloproteinases and their tissue inhibitors in colon mucosa of patients with inflammatory bowel disease. Gut. 2000;47:63-73.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Kobayashi K, Arimura Y, Goto A, Okahara S, Endo T, Shinomura Y, Imai K. Therapeutic implications of the specific inhibition of causative matrix metalloproteinases in experimental colitis induced by dextran sulphate sodium. J Pathol. 2006;209:376-383.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Gordon JN, Pickard KM, Di Sabatino A, Prothero JD, Pender SL, Goggin PM, MacDonald TT. Matrix metalloproteinase-3 production by gut IgG plasma cells in chronic inflammatory bowel disease. Inflamm Bowel Dis. 2008;14:195-203.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Louis E, Ribbens C, Godon A, Franchimont D, De Groote D, Hardy N, Boniver J, Belaiche J, Malaise M. Increased production of matrix metalloproteinase-3 and tissue inhibitor of metalloproteinase-1 by inflamed mucosa in inflammatory bowel disease. Clin Exp Immunol. 2000;120:241-246.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Vaalamo M, Karjalainen-Lindsberg ML, Puolakkainen P, Kere J, Saarialho-Kere U. Distinct expression profiles of stromelysin-2 (MMP-10), collagenase-3 (MMP-13), macrophage metalloelastase (MMP-12), and tissue inhibitor of metalloproteinases-3 (TIMP-3) in intestinal ulcerations. Am J Pathol. 1998;152:1005-1014.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Morgan AR, Han DY, Lam WJ, Triggs CM, Fraser AG, Barclay M, Gearry RB, Meisner S, Stokkers P, Boeckxstaens GE. Genetic variations in matrix metalloproteinases may be associated with increased risk of ulcerative colitis. Hum Immunol. 2011;72:1117-1127.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Maclachlan I. siRNAs with guts. Nat Biotechnol. 2008;26:403-405.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Peer D, Park EJ, Morishita Y, Carman CV, Shimaoka M. Systemic leukocyte-directed siRNA delivery revealing cyclin D1 as an anti-inflammatory target. Science. 2008;319:627-630.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Li MC, He SH. IL-10 and its related cytokines for treatment of inflammatory bowel disease. World J Gastroenterol. 2004;10:620-625.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Correa I, Veny M, Esteller M, Piqué JM, Yagüe J, Panés J, Salas A. Defective IL-10 production in severe phenotypes of Crohn's disease. J Leukoc Biol. 2009;85:896-903.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Santaolalla R, Mañé J, Pedrosa E, Lorén V, Fernández-Bañares F, Mallolas J, Carrasco A, Salas A, Rosinach M, Forné M. Apoptosis resistance of mucosal lymphocytes and IL-10 deficiency in patients with steroid-refractory Crohn's disease. Inflamm Bowel Dis. 2011;17:1490-1500.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Herfarth H, Schölmerich J. IL-10 therapy in Crohn's disease: at the crossroads. Treatment of Crohn's disease with the anti-inflammatory cytokine interleukin 10. Gut. 2002;50:146-147.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Buruiana FE, Solà I, Alonso-Coello P. Recombinant human interleukin 10 for induction of remission in Crohn's disease. Cochrane Database Syst Rev. 2010;CD005109.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Wang AH, Lam WJ, Han DY, Ding Y, Hu R, Fraser AG, Ferguson LR, Morgan AR. The effect of IL-10 genetic variation and interleukin 10 serum levels on Crohn's disease susceptibility in a New Zealand population. Hum Immunol. 2011;72:431-435.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Bhavsar MD, Amiji MM. Oral IL-10 gene delivery in a microsphere-based formulation for local transfection and therapeutic efficacy in inflammatory bowel disease. Gene Ther. 2008;15:1200-1209.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Wirtz S, Neurath MF. Mouse models of inflammatory bowel disease. Adv Drug Deliv Rev. 2007;59:1073-1083.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, Nusrat A. Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol. 2003;171:6164-6172.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Allison MC, Cornwall S, Poulter LW, Dhillon AP, Pounder RE. Macrophage heterogeneity in normal colonic mucosa and in inflammatory bowel disease. Gut. 1988;29:1531-1538.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Seldenrijk CA, Drexhage HA, Meuwissen SG, Pals ST, Meijer CJ. Dendritic cells and scavenger macrophages in chronic inflammatory bowel disease. Gut. 1989;30:484-491.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Ulbrich W, Lamprecht A. Targeted drug-delivery approaches by nanoparticulate carriers in the therapy of inflammatory diseases. J R Soc Interface. 2010;7 Suppl 1:S55-S66.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Laroui H, Wilson DS, Dalmasso G, Salaita K, Murthy N, Sitaraman SV, Merlin D. Nanomedicine in GI. Am J Physiol Gastrointest Liver Physiol. 2011;300:G371-G383.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Mohanraj VJ, Chen Y. Nanoparticles–A Review. Trop J Pharm Res. 2006;5:561-573.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  des Rieux A, Ragnarsson EG, Gullberg E, Préat V, Schneider YJ, Artursson P. Transport of nanoparticles across an in vitro model of the human intestinal follicle associated epithelium. Eur J Pharm Sci. 2005;25:455-465.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Seifert J, Sass W. Intestinal absorption of macromolecules and small particles. Dig Dis. 1990;8:169-178.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Cuthbert AP, Fisher SA, Mirza MM, King K, Hampe J, Croucher PJ, Mascheretti S, Sanderson J, Forbes A, Mansfield J. The contribution of NOD2 gene mutations to the risk and site of disease in inflammatory bowel disease. Gastroenterology. 2002;122:867-874.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Ahmad T, Armuzzi A, Bunce M, Mulcahy-Hawes K, Marshall SE, Orchard TR, Crawshaw J, Large O, de Silva A, Cook JT. The molecular classification of the clinical manifestations of Crohn's disease. Gastroenterology. 2002;122:854-866.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Volkheimer G. [Persorption of microparticles]. Pathologe. 1993;14:247-252.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Hillyer JF, Albrecht RM. Gastrointestinal persorption and tissue distribution of differently sized colloidal gold nanoparticles. J Pharm Sci. 2001;90:1927-1936.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Zabirnyk O, Yezhelyev M, Seleverstov O. Nanoparticles as a novel class of autophagy activators. Autophagy. 2007;3:278-281.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Pertuit D, Moulari B, Betz T, Nadaradjane A, Neumann D, Ismaïli L, Refouvelet B, Pellequer Y, Lamprecht A. 5-amino salicylic acid bound nanoparticles for the therapy of inflammatory bowel disease. J Control Release. 2007;123:211-218.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Moulari B, Pertuit D, Pellequer Y, Lamprecht A. The targeting of surface modified silica nanoparticles to inflamed tissue in experimental colitis. Biomaterials. 2008;29:4554-4560.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Lamprecht A, Yamamoto H, Takeuchi H, Kawashima Y. Nanoparticles enhance therapeutic efficiency by selectively increased local drug dose in experimental colitis in rats. J Pharmacol Exp Ther. 2005;315:196-202.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Meissner Y, Pellequer Y, Lamprecht A. Nanoparticles in inflammatory bowel disease: particle targeting versus pH-sensitive delivery. Int J Pharm. 2006;316:138-143.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Laroui H, Dalmasso G, Nguyen HT, Yan Y, Sitaraman SV, Merlin D. Drug-loaded nanoparticles targeted to the colon with polysaccharide hydrogel reduce colitis in a mouse model. Gastroenterology. 2010;138:843-53.e1-2.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Nakase H, Okazaki K, Tabata Y, Chiba T. Biodegradable microspheres targeting mucosal immune-regulating cells: new approach for treatment of inflammatory bowel disease. J Gastroenterol. 2003;38 Suppl 15:59-62.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Serpe L, Canaparo R, Daperno M, Sostegni R, Martinasso G, Muntoni E, Ippolito L, Vivenza N, Pera A, Eandi M. Solid lipid nanoparticles as anti-inflammatory drug delivery system in a human inflammatory bowel disease whole-blood model. Eur J Pharm Sci. 2010;39:428-436.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Nel AE, Mädler L, Velegol D, Xia T, Hoek EM, Somasundaran P, Klaessig F, Castranova V, Thompson M. Understanding biophysicochemical interactions at the nano-bio interface. Nat Mater. 2009;8:543-557.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Harding CV, Song R. Phagocytic processing of exogenous particulate antigens by macrophages for presentation by class I MHC molecules. J Immunol. 1994;153:4925-4933.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Lamprecht A, Schäfer U, Lehr CM. Size-dependent bioadhesion of micro- and nanoparticulate carriers to the inflamed colonic mucosa. Pharm Res. 2001;18:788-793.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Paulo CS, Pires das Neves R, Ferreira LS. Nanoparticles for intracellular-targeted drug delivery. Nanotechnology. 2011;22:494002.  [PubMed]  [DOI]  [Cited in This Article: ]