Translational Medicine

Translational Medicine
Open Access

ISSN: 2161-1025

Review Article - (2012) Volume 0, Issue 0

Clinical Studies of Molecular Targeted Therapies for Inflammatory Bowel Disease

Kazuhiko Uchiyama, Tomohisa Takagi and Yuji Naito*
Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, Japan
*Corresponding Author: Yuji Naito, Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho Hirokoji Kawaramachi Kamigyo-ku, Kyoto, Japan, Tel: +81-75-251-5519, Fax: +81-75-251-0710 Email:

Abstract

Therapeutic strategies for inflammatory bowel disease (IBD), particularly biological therapies, have been developed recently. The pathogenesis of IBD is based on complicated cytokine-mediated signaling pathways, which represent future drug targets. Recent data have shown that these pathways induce intestinal T-cell activation, which is a central process in disease pathogenesis, via inflammatory mediators. These inflammatory mediators, including cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-12/IL-23, and IL-10, may play important roles in disease pathogenesis, and therefore represent potential therapeutic targets. These strategies may lead to new therapeutic drugs that are more effective and less toxic in IBD.

<

Keywords: Ulcerative colitis, Crohn?s disease, Molecular target, T cell

Introduction

Inflammatory bowel disease (IBD) is defined as chronically relapsing inflammations of the gastrointestinal tract that are not caused by identified pathogens [1,2]. IBD is generally thought to develop as a result of an abnormally active intestinal immune system that results from host-microbial interactions in a genetically susceptible individual [3]. Ulcerative colitis (UC) and Crohn’s disease (CD) are the 2 major forms of IBD that markedly influence the health-related quality of life in patients and are expensive to manage[ 4-6]. UC is characterized by a superficial, continuous inflammation, which is limited to the large intestine, and its symptoms are primarily diarrhea with bleeding. CD is a multifocal, transmural inflammatory process that can affect any part of the digestive tract, and is complicated by deep ulcerations that have the potential to form fistulae and abscesses. Infectious agents, environmental factors, genetic predisposition, and mucosal imbalance with ongoing activation of the intestinal immune system have been implicated in the pathogenesis of IBD [7]. Interestingly, some genes are common to both disorders, suggesting that a part of CD and UC have common genetic and pathophysiologic mechanisms of illness and are not 2 distinct disease entities.

As the exact pathogenic mechanisms are still incompletely understood, therapeutic strategies have been limited to mostly evidencebased principles. Corticosteroids and immunosuppressive agents such asazathioprine, which ameliorate nonspecific inflammatory processes, have been used as conventional treatment for IBD. However, fora majority of IBD patients, corticosteroids are insufficient treatment, as population-based studies have shown that a significant proportion of corticosteroid-treated CD patients develop steroid dependency or even steroid-refractory illness [8], while the need for surgical intervention has apparently remained unchanged [9]. This insufficient therapeutic effect and the potentially severe side effects of corticosteroids have demonstrated the necessity for a more specific therapeutic response, which can only result from a more comprehensive approach of targeting critical points in the signal transduction pathways involved in the inflammatory cascade [10]. Several factors, such as genetic and immunologic abnormalities, are involved in the pathogenesis of IBD. In the context of environmental triggers, these factors lead to the common end result of increased intestinal inflammation and provide multiple potential targets for pharmacotherapeutic intervention. These targets include aberrant host-microbial interactions at the luminalepithelial- cell interface, antigen processing by macrophages and antigen presentation to T cells, T-cell activation, T-cell signaling and T-cell differentiation, cytokine production, and leukocyte trafficking (Figure 1).

translational-medicine-therapeutic

Figure 1: Therapeutic targets for ulcerative colitis and Crohn’s disease. IBD is included these complicated immunologically driven interactions. These cascades might be targets for biologic therapies.

Recent understanding of immunopathogenic mechanisms in IBD has led to the development of biological therapies, which selectively inhibit crucial mediators of the inflammatory process. There is a conception that uncontrolled activation of central effector cells in the gut is the pivotal pathogenic mechanism underlying the initiation and perpetuation of the inflammatory reaction [11,12]. In IBD, CD4+ T cells have been reported to be the main type of activated immune cells, characterized by enhanced proliferation and trafficking into the intestinal mucosa [13]. Alterations in cytokine production elicited by this aberrant T-cell activation lead to a disturbed balance between proand anti-inflammatory cytokines [14]. Profiles of these cytokine are currently the main focus of not only basic but also clinical research in IBD because of the development of new therapeutic strategies.

Tumor Necrosis Factor-α (TNF-α)

In IBD, activated macrophages and lymphocytes produce various pro inflammatory cytokines. One of them, TNF-α plays an important role in IBD. TNF-α exists as a transmembrane protein (membranebound TNF [mTNF]), from which the soluble form (sTNF) is released via proteolytic cleavage by the TNF-α-converting enzyme, and secreted TNF-αexerts its biological functions via 2 distinct cell surface receptors, TNF receptor 1 (TNFR1) and TNFR2 [15,16]. Several studies have demonstrated that the production of TNF-α is elevated in the intestinal mucosa and serum of IBD patients [17-19]. Therefore, this cytokine has been reported to be an important target for the treatment of IBD, and several anti-TNF-α agents have been developed. Among them, infliximab, adalimumab, and certolizumabpegol have proven their therapeutic effects in several studies in CD patients [20- 25]. The efficacy of anti-TNF-α agents is based on multiple effects, but the precise molecular mechanism of action is still unclear. It has been reported that infliximab can block both soluble TNF-α and mTNF-α in vitro [22]. Given that the pathogenesis of CD depends more on effects mediated via mTNF-α, the neutralization of soluble TNF-α is not the only therapeutic modality of anti-TNF-α agents [26]. Several studies on the mechanism of action of infliximab in IBD have demonstrated that it induces apoptosis in a Fas-independent manner via caspase activation in circulating peripheral blood monocytes from CD patients. Mitochondrial release of cytochrome c and transcriptional activation of the proapoptotic proteins Bax and Bakhave also been reported [27]. The apoptotic effect of infliximab on lamina propria T cells at the site of intestinal inflammation was demonstrated in biopsy samples [28]. Intestinal biopsies were taken from macroscopically inflamed areas in CD patients before and 24h after a single infusion of infliximab, and T-cell apoptosis was examined. Infliximab also induced apoptosis in a Fas-independent manner in cultured intestinal T cells from CD patients [29,30]. These data suggest that anti-TNF-α antibodies induce apoptosis in lamina propria mononuclear cells, explaining their clinical efficacy in the treatment of CD. Van den Brande et al. [31] demonstrated the apoptotic effect of infliximab in the lamina propria T cells of patients with active CD and showed that induction of intestinal T-cell apoptosis was related to the clinical efficacy of anti-TNF-α treatment in these patients. These data substantiate the hypothesis that induction of intestinal T-cell apoptosis is one of the central molecular mechanisms of action of anti-TNF-α agents in IBD, and that this effect also explains the clinically visible rapid induction of remission after anti-TNF-α antibody administration. However, these data are not enough to explain the molecular mechanism of action of anti-TNF-α antibodies in IBD because the clinically effective anti-TNF-α agent certolizumab does not induce apoptosis in intestinal T cells.

TNF-α antibody treatment has been reported to contribute colonic mucosal healing of UC and CD patients. Schnitzler et al. [32] reported that long-term maintenance of TNF-α antibody treatment induce mucosal healing and is associated with the avoidance for major abdominal surgeries. Study of Biologic and Immunomodulator Naive Patients in Crohn’s Disease (SONIC Study) revealed that the treatment with infliximab-based strategies resulted in significantly higher rates of mucosal healing at week 26 among patients with active Crohn’s disease [33]. Recent study showed the effect of infliximab about mucosal healing with UC patients [34,35].

TNF-α antibody treatment is the standard strategy to control active CD and UC. However, a large proportion of refractory IBD patients do not respond to TNF-α antibody treatment and the long-term use of these agents are hampered by immunogenicity and the risk of severe infectious complications. Therefore, for specific groups or subgroups of IBD patients, alternative compounds will be necessary to control intestinal inflammation.

Interleukin-6 (IL-6)

It has been reported that the proinflammatory cytokine IL-6 plays an important role in the pathogenesis of IBD. Serum IL-6 levels were substantially elevated in patients with active CD [36-38]. Given that the concentration of IL-6 is related to the activity of CD, serum IL-6 levels are considered to be a clinically relevant parameter [39]. IL-6 mRNA levels are also elevated in the intestinal mucosa of CD patients [40]. In addition, intestinal IL-6 production highly correlates with the severity of endoscopic and histopathologicsigns of inflammation in CD [41,42]. Other studies showed that the main producers of IL-6 in intestinal mucosa are lamina propria T cells [43] and macrophages [44]. In studies on a murine model of dextran sulfate sodium-induced colitis, IL-6-deficient mice showed reduced signs of intestinal inflammation [45]. These results indicate that IL-6 plays an important role in the pathogenesis of IBD.

IL-6 exerts its effects by binding to a soluble form of its corresponding receptor (sIL-6R), which is generated by limited proteolysis of the membrane-bound form on the surface of macrophages [46]. An antibody against IL-6R was tested in diverse murine models of chronic intestinal inflammation. The inflammatory activity was reduced in all models by anti-IL-6R antibody treatment, thereby confirming the role of IL-6-mediated trans-signaling in mucosal inflammation in vivo [43,47]. The anti-IL-6R antibody has been tested in a pilot clinical study [48,49] and analysis of CDAI scores revealed that the antibody was effective in active CD patients compared to the placebo control. However, endoscopic and histologic scores revealed no differences between the anti-IL-6R antibody and placebo groups, suggesting that there is no effect of this antibody on mucosal healing.

IL-12/IL-23

Genome-wide association scans have identified more than 50 susceptibility genes and protective genes for CD and several genes for UC [50-52]. Many of these genes encode proteins associated with inflammatory pathways. Multiple genetic variants in the IL-12/IL-23 pathway that are already known to be associated with CD have been identified by a novel gene pathway analysis [53]. IL-12 and IL-23 are structurally similar heterodimeric cytokines that share a common subunit, p40 (the product of IL-12B). Multiple stages of the immune response and differentiation of naïve CD4+ T cells are modulated by these cytokines. IL-12 promotes the differentiation of CD4+ cells into Th1 cells, which secrete IFN-γ and TNF-α; IL-23 enables the differentiation of CD4+ cells into Th17 cells, which are important in inflammatory regulation [3]. Therefore, the IL-12/IL-23 pathway and the IL-17 pathway are targeted for early phase clinical trials in both CD and UC. Antibodies against IL-12, p40, IL-17 receptor, JAK3, and IL-23 receptor are included in the strategy to control IBD. Antibodies against IL-12 and IL-23 have been evaluated in early phase II studies and these agents have demonstrated efficacy in subpopulations of patients with CD. In a phase III study, ustekinumab, which is a parenteral formulation of IL-12 and IL-23 antibodies and targets the

p40 subunit common to both IL-12 and IL-23, was shown to be highly effective in the treatment of patients with psoriasis [54]. The phase II study of ustekinumabin moderate-to-severe CD patients demonstrated efficacy primarily in patients who had previously received the anti- TNF-α antibody infliximab [26]. Another anti-IL-12/anti-IL-23 antibody apilimodmesylate is an oral compound that suppresses the transcription of IL-12 and IL-23. However, a phase II randomized controlled study showed that this compound was not more effective than placebo for the treatment of active CD [55].

IL-10

A European genome-wide association study identified IL-10 genevariants as susceptibility genes for UC [51]. The anti-inflammatory cytokine IL-10 prevents intestinal inflammation, and genetically deficient IL-10 knockout mice spontaneously develop colitis [56]. Based on these observations, subcutaneous administration of IL-10 was evaluated in CD clinical trials. AlthoughIL-10 administration was not effective in CD patients [57], this drug should be evaluated for UC treatment; an early phase clinical trial is currently underway for the assessment of this product both as an oral pill and a rectal enema formulation.

INF-γ

In patients with CD, it has been reported that the production of IL-12 and IL-18 results in a Th1 polarized immune response, which is thought to be one of the important processes in the pathogenesis of CD. Therefore, it is reasonable to treat CD using agents that block INF-γ, a representative Th1 cytokine. In a CD clinical trial evaluating fontolizumab, an anti-INF-γ-antibody, the active CD group comprising 201 patients exhibited greater improvement in CD activity index scores and C-reactive protein (CRP) levels compared to the placebo group [58]. Although a strong clinical response to fontolizumab in CD was not observed in that phase II study, CRP levels in fontolizumab-treated CD patients were significantly reduced. Two other clinical trials of fontolizumab were conducted on 133 and 45 active CD patients, respectively [59,60]. These studies also demonstrated higher rates of clinical response and remission in the fontolizumab groups compared to the placebo groups. Given the bioactivity of the INF-γ antibody in CD treatment, further large scale clinical trials will be necessary to establish clinical efficacy in CD.

Toll-like Receptors (TLRs)

All experimental animal colitis models require enteric bacteria for the development of intestinal inflammation, and fecal diversion has been accepted as a treatment for distal intestinal inflammation [61]. The recognition of various pathogen-associated molecular patterns, such as lipopolysaccharide, peptidoglycan motifs, and bacterial flagellins, are included in the innate immune system between intestinal flora and intestinal epithelial cells [62]. Toll-like receptors (TLRs) recognize and respond to these bacterial components and induce various intracellular cascades, such as proinflammatory cytokine production [63]. In colonic inflammation, the expression of TLRs on colonic epithelial cells are abnormal, and genetic variants in TLR2, TLR4, TLR5, and TLR9 have been reported to be associated with CD [64-67]. TLR4 induces continuous proinflammatory cytokine production in the presence of Gram-negative bacteria and is up regulated in the colonic mucosa of IBD patients [68]. TLRs are essential for host-microbe interactions and represent potential targets for future biological therapies to limit such interactions. Currently, there are no active trials using TLR agonists or antagonists to disrupt aberrant bacterial-host interactions in IBD patients.

Adhesion Molecules

Several endothelial adhesion molecules, such as E-select in, ICAM- 1, ICAM-2, VCAM-1, and the mucosal address in Mad-CAM-1, could enhance inflammation by modulating leukocyte trafficking and recruiting immune cells into the gut. These molecules interact with integrins on leukocytes, inducting their migration from blood vessels into the site of inflammation in the pathogenesis of IBD. Some of these adhesion molecules represent attractive targets for the development of new drugs for reducing and preventing the recurrence of inflammation, and for long-term control of disease [69]. A selective inhibitor of the human α4-subunit, natalizumab, which inhibits both the VCAM-1/ α4β1 and MAdCAM-1/α4β7 pathways of leukocyte adhesion and transmigration, respectively, has been approved for the treatment of patients with moderate-to-severe CD. The therapeutic effect of natalizumab was superior to that of placebo, demonstrating early and sustained efficacy of the drug as induction therapy in patients with active CD [70]. The monoclonal antibody MLN-02 recognizes α4β7 integrin and selectively inhibits leukocyte adhesion in the gastrointestinal mucosa. The clinical study showed higher clinical remission rates at day 57 (53%) in active CD patients in the MLN-02 group compared to the placebo group, suggesting a beneficial effect of the drug on clinical remission [71]. MLN-02 has also been shown to have beneficial effects in UC patients [72]. Anti-ICAM-1 therapy, using an antisense ICAM-1 (CD54) oligonucleotide that is specifically designed to inhibit ICAM-1 expression, could be useful in the treatment of IBD patients. Although systemic treatment in CD patients revealed no significant results, topical application in the form of enemas has demonstrated some effect in secondary outcomes, and initial studies in pouch it is are promising [73].

Growth Factors

The downstream effects of human growth factors are associated with cellular functions, including epithelial healing in response to injury [74]. As previously reported, impaired epithelial repair is an important pathophysiological event in IBD. Several growth factors, such as epidermal growth factor, keratinocyte growth factor, growth hormone, teduglutide, and GM-CSF/G-CSF have emerged as potential tools for the modulation of intestinal inflammation and tissue repair. GM-CSF did not reveal benefits over placebo in patients treated with 5-ASA; however, steroid-dependent CD patient sexhibiteda higher rate of steroid-free remission compared to patients on placebo [75]. G-CSF has been shown to prevent postoperative recurrence of CD [76,77]. However, evidence relating to other growth factor treatments in CD patients is limited. The phase II trials of keratinocyte growth factor-2 and epidermal growth factor, in particular, did not show efficacy in CD or UC patients [78,79].

Conclusion

The pathogenesis of IBD is complicated and therapeutic target cells include not only immune cells, such as T cells and macrophages, but also colonic epithelial cells. A number of molecules are also associated with the pathogenesis of IBD. Increased understanding of the immune mechanisms in IBD has facilitated the development of clinically effective biologics target such molecules. Future biological agents, therefore, carry an enormous potential for optimized therapeutic efficacy in patients with IBD.

References

  1. Podolsky DK (1991) Inflammatory bowel disease (2). N Engl J Med 325: 1008-1016.
  2. Podolsky DK (1991) Inflammatory bowel disease (1). N Engl J Med 325:928-37.
  3. Shih DQ, Targan SR (2008) Immunopathogenesis of inflammatory bowel disease. World J Gastroenterol 14: 390-400.
  4. Drossman DA, Patrick DL, Mitchell CM, Zagami EA, Appelbaum MI (1989) Health-related quality of life in inflammatory bowel disease. Functional status and patient worries and concerns. Dig Dis Sci 34: 1379-1386.
  5. Kappelman MD, Rifas-Shiman SL, Kleinman K, Ollendorf D, Bousvaros A, et al. (2007) The prevalence and geographic distribution of Crohn's disease and ulcerative colitis in the United States. Clin Gastroenterol Hepatol 5: 1424-1429.
  6. Kappelman MD, Rifas-Shiman SL, Porter CQ, Ollendorf DA, Sandler RS, et al. (2008) Direct health care costs of Crohn's disease and ulcerative colitis in US children and adults. Gastroenterology 135: 1907-1913.
  7. Podolsky DK (2002) Inflammatory bowel disease. N Engl J Med 347: 417-429.
  8. Munkholm P, Langholz E, Hollander D, Thornberg K, Orholm M, et al. (1994) Intestinal permeability in patients with Crohn's disease and ulcerative colitis and their first degree relatives. Gut 35: 68-72.
  9. Cosnes J, Nion-Larmurier I, Beaugerie L, Afchain P, Tiret E, et al. (2005) Impact of the increasing use of immunosuppressants in Crohn's disease on the need for intestinal surgery. Gut 54: 237-241.
  10. Sartor RB (1994) Cytokines in intestinal inflammation: pathophysiological and clinical considerations. Gastroenterology 106: 533-539.
  11. Neurath MF, Finotto S, Fuss I, Boirivant M, Galle PR, et al. (2001) Regulation of T-cell apoptosis in inflammatory bowel disease: to die or not to die, that is the mucosal question. Trends Immunol 22: 21-26
  12. Fiocchi C (1998) Inflammatory bowel disease: etiology and pathogenesis. Gastroenterology 115: 182-205.
  13. Fuss IJ, Neurath M, Boirivant M, Klein JS, de la Motte C, et al. (1996) Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn's disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol 157: 1261-1270.
  14. Eissner G, Kolch W, Scheurich P (2004) Ligands working as receptors: reverse signaling by members of the TNF superfamily enhance the plasticity of the immune system. Cytokine Growth Factor Rev 15: 353-366.
  15. Wajant H, Pfizenmaier K, Scheurich P (2003)Tumor necrosis factor signaling. Cell Death Differ 10: 45-65.
  16. Reimund JM, Wittersheim C, Dumont S, Muller CD, Baumann R, et al. (1996) Mucosal inflammatory cytokine production by intestinal biopsies in patients with ulcerative colitis and Crohn's disease. J Clin Immunol 16: 144-150.
  17. Breese EJ, Michie CA, Nicholls SW, Murch SH, Williams CB, et al. (1994) Tumor necrosis factor alpha-producing cells in the intestinal mucosa of children with inflammatory bowel disease. Gastroenterology 106: 1455-1466.
  18. Murch SH, Braegger CP, Walker-Smith JA, MacDonald TT (1993) Location of tumour necrosis factor alpha by immunohistochemistry in chronic inflammatory bowel disease. Gut 34: 1705-1709.
  19. Targan SR, Hanauer SB, van Deventer SJ, Mayer L, Present DH, et al. (1997) A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn's disease. Crohn's Disease cA2 Study Group. N Engl J Med 337: 1029-1035.
  20. Hanauer SB, Feagan BG, Lichtenstein GR, Mayer LF, Schreiber S, et al. (2002) Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial. Lancet 359: 1541-1549.
  21. Hanauer SB, Sandborn WJ, Rutgeerts P, Fedorak RN, Lukas M, et al. (2006) Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology 130: 323-333.
  22. Colombel JF, Sandborn WJ, Rutgeerts P, Enns R, Hanauer SB, et al. (2007) Adalimumab for maintenance of clinical response and remission in patients with Crohn's disease: the CHARM trial. Gastroenterology 132: 52-65
  23. Sandborn WJ, Feagan BG, Stoinov S, Honiball PJ, Rutgeerts P, et al. (2007) Certolizumab pegol for the treatment of Crohn's disease. N Engl J Med 357: 228-238
  24. Schreiber S, Khaliq-Kareemi M, Lawrance IC, Thomsen OØ, Hanauer SB, et al. (2007) Maintenance therapy with certolizumab pegol for Crohn's disease. N Engl J Med 357: 239-250.
  25. Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR, et al. (2008) A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology 135: 1130-1141.
  26. Lügering A, Schmidt M, Lügering N, Pauels HG, Domschke W, et al. (2001) Infliximab induces apoptosis in monocytes from patients with chronic active Crohn's disease by using a caspase-dependent pathway. Gastroenterology 121: 1145-1157.
  27. ten Hove T, van Montfrans C, Peppelenbosch MP, van Deventer SJ (2002) Infliximab treatment induces apoptosis of lamina propria T lymphocytes in Crohn's disease. Gut 50: 206-211.
  28. Di Sabatino A, Ciccocioppo R, Cinque B, Millimaggi D, Morera R, et al. (2004) Defective mucosal T cell death is sustainably reverted by infliximab in a caspase dependent pathway in Crohn's disease. Gut 53: 70-77.
  29. Van den Brande JM, Braat H, van den Brink GR, Versteeg HH, Bauer CA, et al. (2003) Infliximab but not etanercept induces apoptosis in lamina propria T-lymphocytes from patients with Crohn's disease. Gastroenterology 124: 1774-1785.
  30. Van den Brande JM, Koehler TC, Zelinkova Z, Bennink RJ, te Velde AA, et al. (2007) Prediction of antitumour necrosis factor clinical efficacy by real-time visualisation of apoptosis in patients with Crohn's disease. Gut 56: 509-517.
  31. Schnitzler F, Fidder H, Ferrante M, Noman M, Arijs I, et al. (2009) Mucosal healing predicts long-term outcome of maintenance therapy with infliximab in Crohn's disease. Inflamm Bowel Dis 15: 1295-1301.
  32. Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A, et al. (2010) Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med 362: 1383-1395.
  33. Fratila OC, Craciun C (2010) Ultrastructural evidence of mucosal healing after infliximab in patients with ulcerative colitis. J Gastrointestin Liver Dis 19: 147-153.
  34. Barreiro-de Acosta M, Lorenzo A, Mera J, Dominguez-Muñoz JE (2009) Mucosal healing and steroid-sparing associated with infliximab for steroid-dependent ulcerative colitis. J Crohns Colitis 3: 271-276.
  35. Gross V, Andus T, Caesar I, Roth M, Schölmerich J (1992) Evidence for continuous stimulation of interleukin-6 production in Crohn's disease. Gastroenterology102: 514-519.
  36. Holub MC, Makó E, Dévay T, Dank M, Szalai C, et al. (1998) Increased interleukin-6 levels, interleukin-6 receptor and gp130 expression in peripheral lymphocytes of patients with inflammatory bowel disease. Scand J Gastroenterol Suppl 228: 47-50.
  37. Hyams JS, Fitzgerald JE, Treem WR, Wyzga N, Kreutzer DL (1993) Relationship of functional and antigenic interleukin 6 to disease activity in inflammatory bowel disease. Gastroenterology 104: 1285-1292.
  38. Reinisch W, Gasché C, Tillinger W, Wyatt J, Lichtenberger C, et al. (1999) Clinical relevance of serum interleukin-6 in Crohn's disease: single point measurements, therapy monitoring, and prediction of clinical relapse. Am J Gastroenterol 94: 2156-2164
  39. Isaacs KL, Sartor RB, Haskill S (1992) Cytokine messenger RNA profiles in inflammatory bowel disease mucosa detected by polymerase chain reaction amplification. Gastroenterology 103:1587-1595.
  40. Reinecker HC, Steffen M, Witthoeft T, Pflueger I, Schreiber S, et al. (1993) Enhanced secretion of tumour necrosis factor-alpha, IL-6, and IL-1 beta by isolated lamina propria mononuclear cells from patients with ulcerative colitis and Crohn's disease. Clin Exp Immunol 94: 174-181.
  41. Kusugami K, Fukatsu A, Tanimoto M, Shinoda M, Haruta J, et al. (1995) Elevation of interleukin-6 in inflammatory bowel disease is macrophage- and epithelial cell-dependent. Dig Dis Sci 40: 949-959.
  42. Atreya R, Mudter J, Finotto S, Müllberg J, Jostock T, et al. (2000) Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo. Nat Med 6: 583-588
  43. Taga T, Hibi M, Hirata Y, Yawata H, Natsuka S, et al. (1989) Interleukin-6 receptor and a unique mechanism of its signal transduction. Cold Spring Harb Symp Quant Biol 2: 713-722.
  44. Suzuki A, Hanada T, Mitsuyama K, Yoshida T, Kamizono S, et al. (2001) CIS3/SOCS3/SSI3 plays a negative regulatory role in STAT3 activation and intestinal inflammation. J Exp Med 193: 471-481.
  45. Mitsuyama K, Toyonaga A, Sasaki E, Ishida O, Ikeda H, et al. (1995) Soluble interleukin-6 receptors in inflammatory bowel disease: relation to circulating interleukin-6. Gut 36: 45-49.
  46. Yamamoto M, Yoshizaki K, Kishimoto T, Ito H (2000) IL-6 is required for the development of Th1 cell-mediated murine colitis. J Immunol 164: 4878-4882.
  47. Ito H, Takazoe M, Fukuda Y, Hibi T, Kusugami K, et al. (2004) A pilot randomized trial of a human anti-interleukin-6 receptor monoclonal antibody in active Crohn's disease. Gastroenterology 126: 989-996.
  48. Ito H (2005) Treatment of Crohn's disease with anti-IL-6 receptor antibody. J Gastroenterol 16: 32-34.
  49. Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, et al. (2008) Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet 40: 955-962.
  50. Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, et al. (2008) Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat Genet 40: 1319-1323.
  51. Silverberg MS, Cho JH, Rioux JD, McGovern DP, Wu J, et al. (20069) Ulcerative colitis-risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study. Nat Genet 41: 216-220.
  52. Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, et al. (2006) A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314: 1461-1463.
  53. Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, et al. (2008) Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 371: 1675-1684.
  54. Sands BE, Jacobson EW, Sylwestrowicz T, Younes Z, Dryden G, et al. (2010) Randomized, double-blind, placebo-controlled trial of the oral interleukin-12/23 inhibitor apilimod mesylate for treatment of active Crohn's disease. Inflamm Bowel Dis 16: 1209-1218.
  55. Berg DJ, Davidson N, Kühn R, Müller W, Menon S ,et al. (1996) Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest 98: 1010-1020.
  56. Schreiber S, Fedorak RN, Nielsen OH, Wild G, Williams CN, et al. (2000) Safety and efficacy of recombinant human interleukin 10 in chronic active Crohn's disease. Crohn's Disease IL-10 Cooperative Study Group. Gastroenterology 119: 1461-1472.
  57. Reinisch W, de Villiers W, Bene L, Simon L, Rácz I, et al. (2010) Fontolizumab in moderate to severe Crohn's disease: a phase 2, randomized, double-blind, placebo-controlled, multiple-dose study. Inflamm Bowel Dis 16: 233-242.
  58. Hommes DW, Mikhajlova TL, Stoinov S, Stimac D, Vucelic B, et al. (2006) Fontolizumab, a humanised anti-interferon gamma antibody, demonstrates safety and clinical activity in patients with moderate to severe Crohn's disease. Gut 55: 1131-1137.
  59. Reinisch W, Hommes DW, Van Assche G, Colombel JF, Gendre JP, et al. (2006) A dose escalating, placebo controlled, double blind, single dose and multidose, safety and tolerability study of fontolizumab, a humanised anti-interferon gamma antibody, in patients with moderate to severe Crohn's disease. Gut 55: 1138-1144.
  60. Burman JH, Williams JA, Thompson H, Cooke WT (1969) The effect of diversion of intestinal contents on the progress of Crohn's disease of the large bowel. Gut 10: 1054.
  61. Melmed GY, Abreu MT (2004) New insights into the pathogenesis of inflammatory bowel disease. Curr Gastroenterol Rep 6: 474-481.
  62. Rosenstiel P, Till A, Schreiber S (2007) NOD-like receptors and human diseases. Microbes Infect 9: 648-657.
  63. Franchimont D, Vermeire S, El Housni H, Pierik M, Van Steen K, et al. (2004) Deficient host-bacteria interactions in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn's disease and ulcerative colitis. Gut 53: 987-992.
  64. Gewirtz AT, Vijay-Kumar M, Brant SR, Duerr RH, Nicolae DL, et al. (2006) Dominant-negative TLR5 polymorphism reduces adaptive immune response to flagellin and negatively associates with Crohn's disease. Am J Physiol Gastrointest Liver Physiol 290: 1157-1163.
  65. Török HP, Glas J, Tonenchi L, Mussack T, Folwaczny C (2004) Polymorphisms of the lipopolysaccharide-signaling complex in inflammatory bowel disease: association of a mutation in the Toll-like receptor 4 gene with ulcerative colitis. Clin Immunol 112: 85-91
  66. Török HP, Glas J, Tonenchi L, Bruennler G, Folwaczny M, et al. (2004) Crohn's disease is associated with a toll-like receptor-9 polymorphism. Gastroenterology 127: 365-366.
  67. Abreu MT, Arnold ET, Thomas LS, Gonsky R, Zhou Y, et al. (2002) TLR4 and MD-2 expression is regulated by immune-mediated signals in human intestinal epithelial cells. J Biol Chem 277: 20431-20437.
  68. Ghosh S, Panaccione R (2010) Anti-adhesion molecule therapy for inflammatory bowel disease. Therap Adv Gastroenterol 3: 239-258.
  69. Targan SR, Feagan BG, Fedorak RN, Lashner BA, Panaccione R, et al. (2007) Natalizumab for the treatment of active Crohn's disease: results of the ENCORE Trial. Gastroenterology 132: 1672-1683.
  70. Feagan BG, Greenberg GR, Wild G, Fedorak RN, Paré P, et al. Treatment of active Crohn's disease with MLN0002, a humanized antibody to the alpha4beta7 integrin. Clin Gastroenterol Hepatol 6: 1370-1377.
  71. Feagan BG, Greenberg GR, Wild G, Fedorak RN, Paré P, et al. (2005) Treatment of ulcerative colitis with a humanized antibody to the alpha4beta7 integrin. N Engl J Med 352: 2499-2507.
  72. Philpott JR, Miner PB Jr (2008) Antisense inhibition of ICAM-1 expression as therapy provides insight into basic inflammatory pathways through early experiences in IBD. Expert Opin Biol Ther 8: 1627-1632.
  73. Uchiyama K, Naito Y, Takagi T, Mizushima K, Hayashi N, et al. (2011) FGF19 protects colonic epithelial cells against hydrogen peroxide. Digestion 83: 180-183.
  74. Bernasconi E, Favre L, Maillard MH, Bachmann D, Pythoud C, et al. (2010) Granulocyte-macrophage colony-stimulating factor elicits bone marrow-derived cells that promote efficient colonic mucosal healing. Inflamm Bowel Dis 16: 428-441.
  75. Dieckgraefe BK, Korzenik JR (2002) Treatment of active Crohn's disease with recombinant human granulocyte-macrophage colony-stimulating factor. Lancet 360: 1478-1480.
  76. Korzenik JR, Dieckgraefe BK, Valentine JF, Hausman DF, Gilbert MJ, et al. (2005) Sargramostim for active Crohn's disease. N Engl J Med 352: 2193-2201.
  77. Krishnan K, Arnone B, Buchman A (2011) Intestinal growth factors: potential use in the treatment of inflammatory bowel disease and their role in mucosal healing. Inflamm Bowel Dis 17: 410-422.
  78. Barahona-Garrido J, Yamamoto-Furusho JK (2008) New treatment options in the management of IBD - focus on colony stimulating factors. Biologics 2: 501-504.
Citation: Uchiyama K, Takagi T, Naito Y (2012) Clinical Studies of Molecular Targeted Therapies for Inflammatory Bowel Disease. Transl Med S2:004.

Copyright: © 2012 Uchiyama K, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top

https://sekillinickyazma.com.tr/