Review Open Access
Copyright ©2008 The WJG Press and Baishideng. All rights reserved.
World J Gastroenterol. Aug 21, 2008; 14(31): 4861-4866
Published online Aug 21, 2008. doi: 10.3748/wjg.14.4861
Cytokine orchestration in post-operative peritoneal adhesion formation
Ronan A Cahill, H Paul Redmond, Department of General Surgery, Cork University Hospital, Wilton, Cork, Ireland
Author contributions: Cahill RA and Redmond HP contributed equally to the composition of this work.
Supported by Clinical Research Fellowship from the Health Research Board, Ireland
Correspondence to: Ronan A Cahill, Department of General Surgery, Cork University Hospital, Wilton, Cork, Ireland. rcahill@rcsi.ie
Telephone: +353-21-4922373 Fax: +353-21-343307
Received: May 1, 2008
Revised: July 14, 2008
Accepted: July 21, 2008
Published online: August 21, 2008

Abstract

Peritoneal adhesions are a near inevitable occurrence after laparotomy and a major cause of both patient and physician misery. To date, clinical attempts at their amelioration have concentrated on manipulating the physical factors that affect their development despite a wealth of experimental data elucidating the molecular mechanisms that underlie their initiation, development and maturation. However, the advent of targeted, specific anti-cytokine agents as directed therapy for inflammatory and neoplastic conditions raises the prospect of a new era for anti-adhesion strategies. To harness this potential will require considerable cross-disciplinary collaboration and that surgeon-scientists propel themselves to the forefront of this emerging field.

Key Words: Postoperative peritoneal adhesion formation, Cytokines, Vascular endothelial growth factor



INTRODUCTION
Table 1 Overview of literature to date regarding cytokine orchestration in postoperative adhesion formation. Included in the list are cytokines, chemokines, and proteases as well as trigger enzymes.
CytokineRefMechanism investigatedIn vitro/vivoSpeciesExperimental modelEffect on adhesion formation
Heparin-binding growth factor[41]Macrophage and neutrophil omental migrationIn vivoMouse(1) Partial hepatectomy (2) Omental adherenceExacerbated by Midkine- omental inflammation reduced
HGF[42]Mesothelial cell proliferation and migrationBothRatCecal abrasionExacerbated by local HGF gene transfer
IFN-γ , HGF[43]Natural killer T cell activityBothMouseCecal cauterizationAttenuated by HGF
IL-1[44]Nonspecific inflammationIn vivoRatCecal abrasionExacerbated by IL-1
IL-1, TNF[45]Proinflammatory markersIn vivoHumanAdhesion samplesIL-1 & TNF-α associated with adhesion
IL-1, IL-6, TNF-α[46]Cellular mediationIn vitroHumanPeritoneal fluid samplingAdhesions associated with IL-6 and IL-1
IL-10[47]Natural antiiflammatoryIn vivoMousePeritoneal injuryAttenuated by IL-10 but no effect with IL-10 mAb. No associated with IL-10 levels
IL-10[48]ImmunosuppressionIn vivoMousePeritoneal injuryAttenuated by IL-10
IL-1b, TNF-α, TGF-β1, IL-10, IFNg, GM-CSF[49]InflammatoryIn vitroHumanPeritoneal fluid samplingOnly IFN-γ and TGF-β1 associated with adhesion formation. No association found with other cytokines.
IL-6[50]Early proinflammatory effectsIn vivoRatCecal abrasion with C2H5OHExacerbated by IL-6, attenuated by monoclonal Ab to IL-6
PAF[51]Early inflammatory mediatorsIn vivoRatUterine horn abrasionAdhesions and IL-6 levels attenuated by Lexipafant (PAF antagonist)
Substance P[52]Substance P mediationIn vivoRatPeritoneal ischaemic buttonsSubstance P and TGF-β1 as well as ICAM-1 and VCAM-1 increased
TGF[53]TGF isoformsIn vivoMouseSerosal abrasion and appositionExacerbated by TGF-β3, attenuated by combined TGF-β1 and TGF-β2 mAB
TGF-β[54]TGF-β regulation of extracellular matrixIn vivoHumanHuman fibroblast cultureDichloroacetic acid inhibited fibronectin and collagen type III expression
TGF-β[55]ChemoattractionIn vitroRatCecal abrasionTGF-β mRNA increased by trauma
TGF-β[56]Mast cellsIn vivoHamsterUterine horn abrasionExacerbated by chymase inhibitor
TGF-β[57]ChemoattractionIn vivoRatUterine horn abrasionExacerbated by TGF-β
TGF-β[58]Mast cellsIn vitroHumanCell cultureTGF-β and tryptase increased collagen
TGF-β[59]Peritoneal repairIn vivoRatUterine horn abrasionNo antiadhesion effect of anti-TGF mAb
TGF-β[60]ImmunosuppressionIn vivoRatSmall bowel transplantAdhesions attenuated by tacrolimus
TGF-β[61]Mast cellsIn vivoRatUterus scrapingTGF-β increased by trauma, adhesions attenuated by chymase inhibition
TGF-β[62]Cellular effects of TisseelIn vitroHumanCell cultureFibroblasts TGF-β reduced
TGF-β, MMP-9, TIMP-1[63]Matrix factorsIn vivoHumanSampled peritoneal fluidAdhesion assoc with reduced MMP-9 but elevated MMP-9/TIMP-1 ratio
TGF-β/MDF[64]Carboxymethylcellulose spongeIn vivoRatCecal denudation & appositionEffect of sponge independent to cytokine release (barrier function)
TGF-β1[65]ChemoattractionIn vitroHumanCell cultureTGF-β1 increased in scar tissue
TGF-β1[66]Extracellular matrixIn vivoMouseCecal abrasionExacerbated by haploid insufficiency
TGF-β1[67]FibrinolysisIn vitroHumanBiopsy samplingAttenuated by TGF-β1 overexpression
TGF-β1[68]PeritonitisIn vivoRatCecal ligation and puncturePeritonitis upregulates TGF-β1 expression
TGF-β1[69]Mitogenicity of macrophages & fibroblastsIn vivoRatSmall Bowel transection and re-anastomosisAdhesions and TGF-1 levels attenuated by ACE inhibition
TGF-β1, MMP1&2, TPA, TIMP-1[70]Cellular effects of seprafilmIn vitroHumanHuman fibroblast & mesothelial cell cultureNo cytokine effect induced by Seprafilm (barrier effect important)
TGF-β1, TGF-β2[71]Basal expressionIn vitroHumanBiopsy samplingSit-specific TGF-β1 & TGF-β3 expression
TGF-β1[72]Cellular effects of changtongIn vivoRat/rabbitCecal abrasionTGF-β reduced in rats
TNF, IL-1, IL-6[73]Effects of gloves and powdersIn vivoRatCecal abrasionAdhesions increased by glove powder
TNF-α [74]Proinflammatory effects of TNF-αIn vivoRatCecal abrasionAdhesion formation attenuated by infliximab but no histological effect
TNF-α, IL-1[75]Proinflammatory markersIn vivoRatCecal abrasion or small bowel resectionTNF-α appears a good biological marker for adhesion formation
TNF-α, IL-1[76]ImmunosuppressionIn vivoRatCecal abrasionAdhesion formation attenuated by mAbs to IL1 and IL-1/ TNF-α
TNF-α, IL-6[77]Proinflammatory mediatorsIn vitroMouseMurine macrophagesAdhesion formation attenuated by hyaluronic acid and dexamethasone
TNF-α, MMP[78]Mesothelium reaction to peritoneal injuryIn vivoRatPeritoneal woundingNo effect of MMP & TACE inhibition, TNF-α may not be adhesiogenic
TNF-α, TGF-β1[79]PROACT to injured peritoneumIn vivoHumanTissue samplingTNF-α and TGF-β reduced by heating
VEGF[80]AngiogenesisIn vivoRatUterus-peritoneal scrubAssociated by angiogenesis
VEGF[29,32]Vascular permeabilityIn vivoMousePeritoneal injuryAdhesions attenuated by Antiserum and monoclonal antibody
VEGF, basic-FGF[25]Fibrovascular band formationIn vivoHumanAdhesion samplesVEGF in endothelial cells associated with adhesion formation
VEGF, IL-6[21]Bacterial TranslocationBothMouseCaecal abrasion & sutureAdhesions attenuated by rBPI
VEGF, PlGF[81]PnenumoperitoeumIn vivoMouseLap. uterine horn modelExacerbated by VEGF and CO2
CCL 1-CCR 8[83]Specific recruitment of peritoneal macrophagesBothMousePeritoneal ischaemic button & colitis-associated peritoneal adhesionsUnaffected by CCR8 gene deficiency and antiCCL1-neutralizing antibody
CD 28 T cell costimulatory pathway[84]CD28 T cell costimulatory pathway/Inhibitor programmed death-1 pathwayBothMouseCaecal abrasionExacerbated by CD28 T Cell costimulatory pathway but unaffected by death-1 pathway
Interferon-inducible protein-10[85]Regulates influxing neutrophils, monocytes and lymphocytesIn vivoMousePeritoneal side wall injury
Broad spectrum of chemokines[86]Broad spectrum chemokine inhibitor NR58-3.14.3In vivoMousePeritoneal traumatizationAdhesions significantly attenuated
MCP-1[87]Fibroblast and mononuclear cell chemotaxisIn vivoMousePeritoneal injuryAttenuated by MCP-1 antibody
MCP-1[88]Fibroblast and mononuclear cell chemotaxisIn vivoHumanCell culture
MCP-1[89]Fibroblast and mononuclear cell chemotaxisIn vivoHumanCell culture
T cells, IL-17, CXC MPI-2/CXCL8, CXCL1[90]CD4+ T cellsIn vivoMouseCaecal abrasionUnaffected by anti-IL-17 antibodies

Post-operative peritoneal adhesion formation remains a considerable source of patient and physician frustration and a significant burden on hospital resources[1]. As the commonest cause of small bowel obstruction in patients who have previously undergone laparotomy, adhesions account for 40% of all cases of intestinal obstruction and 60%-70% of those affecting the small bowel. After a first such clinical episode, 53% of patients will go on to develop a second relapse, and 83% of these will have chronic symptoms[2]. Some 14% of those who manifest overt adhesive intestinal obstruction do so within 2 years of their initial surgery, with 2.6% requiring operative adhesiolysis for its relief[3]. Furthermore, approximately 20% of patients developing adhesional bowel obstruction do so at a remove of more then ten years after their index operation[4]. Post-operative adhesions are also a common cofactor in female infertility in those with prior laparotomy[5,6] and they add markedly to the technical complexity of any repeat abdominal operation. By doing so, they give rise to considerable surgeon frustration[7] and a heightened risk of patient morbidity[8].

For all these reasons, this iatrogenic complication weighs heavily on the balance books of health care providers. Indeed, in overall costs, the financial cost due to adhesion-related morbidity approximates the expenditure required for the surgical management of gastric or rectal cancer[9] and this is then further compounded by the cost of medicolegal claims and settlements. Finally, the considerable number of bed-days consumed by the sequelae and treatment of post-operative adhesions (indeed in Finland, adhesion-related admissions exceeds the number of bed-days appropriated to varicose vein surgery) also reinforces the urgency for developing effective means of adhesion abrogation.

Unfortunately, however, clinical strategies and therapies aimed at controlling or alleviating adhesion formation have been largely inadequate in their address of both ongoing human suffering[10] and economic cost[11]. To date these attempts have mostly concentrated on employing physical means to align[12-14] or separate[15] adjacent loops of bowel in the early post-operative period (so that any configuration of interloop bands is either organised or hindered respectively) or have focused on manipulating peritoneal fibrinolytic mechanisms[16-18].

CYTOKINE ORCHESTRATION IN POST-OPERATIVE ADHESION FORMATION

Adhesions however represent a form of secondary wound healing. Therefore the mesothelial tissue response to injury (occurring either directly due to handling and dissection or indirectly due to desiccation, cooling or relative ischaemia at sites both adjacent to and distant from the actual operative site) is initiated locally and thence both propagated and orchestrated by cytokine signaling. Although systemic[19] and genetic elements[20] may also influence the severity of the cascade and factors such as bacterial contamination can potentate it[21], interruption or manipulation of key cellular processes early in the response cascade would seem likely to markedly diminish all downstream events including the ultimate fibrotic endpoint. Furthermore, the increasing sophistication of anti-cytokine therapies now allows single components of complex cellular processes to be specifically targeted. In addition, potentially efficacious agents have already been proved both safe and useful in the management of anti-neoplastic[22] and anti-inflammatory conditions[23]. Therefore a new era in the approach to adhesion amelioration may be in the offing.

SPECIFIC TARGETTING OF SELECTED CYTOKINES

There has of course been a vast array of cytokines and chemokines implicated in the initiation, development and maturation of abdominal adhesions after laparotomy (Table 1) and therefore it may initially appear forbidding to try and narrow the therapeutic target most likely to lead to unopposed benefit. Tumor necrosis factor was one of the earliest cytokines investigated and certainly seems to represent one important factor. However its recent elucidation as a key mediator of the bacterial response to infection seems to mitigate against using monoclonal antibodies (already commercially available) to abrogate this cytokine early after intestinal operation[24]. Equally, the variability of action depending on the relative proportions of its isoforms and the central role it plays in wound healing would also seem to deter use of directed therapy against transforming growth factor-beta. Of the remaining candidate targets the majority only really have a slender evidence base to support their selection from out of the general post-operative molecular milieu. The one exception, at present, would seem to be vascular endothelial growth factor (VEGF).

Although this important signaling protein is best known as a potent angiogenic cytokine (and indeed may be proposed as having a role in the process of adhesion growth through the induction of new blood vessels into areas of operative tissue injury[25]), VEGF is now also well established as being directly involved in restorative tissue processes, including early inflammatory responses, as well as wound repair and remodeling via effecting fibroblast function[26]. Furthermore, the central role of VEGF in facilitating increased vascular permeability (essential for the early proinflammatory response to injury) as well as the subsequent deposition of the fibrin-rich matrix necessary for subsequent cellular migration and proliferation[27,28] would seem to make it a prime putative agent in the formation of peritoneal adhesions. It is not surprising therefore that VEGF has been consistently positively implicated (albeit non-selectively) in this process[29]. The realization that peritoneal mast cells both constitutively and inducibly express this cytokine[30,31] further suggests an intriguing link given that these cells are known also to be central to adhesion formation[32]. However, it may well be that rather than through direct secretion, mast cells effect the threshold concentration of this cytokine by exciting the egress of neutrophils and monocytes from the circulation into the peritoneum and that it is these cells that instead then contribute most to regional VEGF levels.

Regardless of its exact cellular origin, VEGF seems to represent an ideal target as its levels correlate with adhesion formation in animal models with its regulation (either positively[19] or negatively[32]) affecting the degree to which they form after peritoneal operations. The clinical success and safety of VEGF neutralization by a specific monoclonal antibody in the treatment of malignant diseases[33] adds further impetus to the need to try its pharmacological manipulation as an anti-adhesion strategy particularly as selective therapeutic targeting of the cytokine does not seem to disrupt operative wound healing in a clinically important fashion[34].

DETERMINATION OF CLINICAL EFFICACY

Clinical evidence of efficacy of anti-adhesion therapies is notoriously difficult to attain as second look-laparotomy to assess distribution and intensity of peritoneal reaction is not ethically justifiable (although may be possible in the case of certain gynecological procedures[35]). Additionally, the mere presence of adhesions, even if extensive, does not necessarily correlate with the incidence and severity of subsequent symptomatic episodes and long-term follow-up is required to determine the full-extent of the problems arising. These challenges are not however insurmountable as have been shown by those who advance the cause of bioactive substances[36,37] and the difficulties that would be encountered in establishing a progressing and adequately powered multi coated blinded study would be markedly outweighed by the huge benefit to patients of many differing specialties. With regard to monoclonal antibody therapies in particular, there now exists the opportunity to piggy-back on the human safety testing performed on this class of drug in alternative settings. While pursuit of molecular mechanisms for adhesion amelioration will undoubtedly still be expensive[38], the cost incurred by the management of adhesion-related morbidity[39,40] economically justifies considerable investment in any potential means of their attenuation.

CONCLUSION

There have long been a multitude of groups proposing novel, potential therapies for the attenuation of adhesion formation at a preclinical level- the onus now though is on leading surgeon-scientists to corral their endeavour and progress their preclinical expertise into the clinical setting. For a start, the most likely candidate cytokine must be agreed (in our mind VEGF would seem the most apposite) and the most appropriate means of affecting its activity (whether directly[32] or indirectly[21]) selected. Furthermore industry interest will need to be stimulated for its support for Phase II and III trials as well as for the subsequent manufacture and marketing processes is crucial. Above all, though it must be realized that the timing for a concerted attempt to prove that molecular manipulation of post-operative peritoneal formation has never been better.

Footnotes

Peer reviewer: Dr. Maria Concepción Gutiérrez-Ruiz, Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Avenida San Rafael Atlixco 186, Colonia Vicentina, Mexico DF 09340, Mexico

S- Editor Zhong XY L- Editor Alpini GD E- Editor Lin YP

References
1.  Ellis H. The clinical significance of adhesions: focus on intestinal obstruction. Eur J Surg Suppl. 1997;5-9.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Barkan H, Webster S, Ozeran S. Factors predicting the recurrence of adhesive small-bowel obstruction. Am J Surg. 1995;170:361-365.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Beck DE, Opelka FG, Bailey HR, Rauh SM, Pashos CL. Incidence of small-bowel obstruction and adhesiolysis after open colorectal and general surgery. Dis Colon Rectum. 1999;42:241-248.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Menzies D, Ellis H. Intestinal obstruction from adhesions--how big is the problem? Ann R Coll Surg Engl. 1990;72:60-63.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Hershlag A, Diamond MP, DeCherney AH. Adhesiolysis. Clin Obstet Gynecol. 1991;34:395-402.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ray NF, Denton WG, Thamer M, Henderson SC, Perry S. Abdominal adhesiolysis: inpatient care and expenditures in the United States in 1994. J Am Coll Surg. 1998;186:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Coleman MG, McLain AD, Moran BJ. Impact of previous surgery on time taken for incision and division of adhesions during laparotomy. Dis Colon Rectum. 2000;43:1297-1299.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Van Der Krabben AA, Dijkstra FR, Nieuwenhuijzen M, Reijnen MM, Schaapveld M, Van Goor H. Morbidity and mortality of inadvertent enterotomy during adhesiotomy. Br J Surg. 2000;87:467-471.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Kossi J, Salminen P, Rantala A, Laato M. Population-based study of the surgical workload and economic impact of bowel obstruction caused by postoperative adhesions. Br J Surg. 2003;90:1441-1444.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Davey AK, Maher PJ. Surgical adhesions: a timely update, a great challenge for the future. J Minim Invasive Gynecol. 2007;14:15-22.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Kossi JA, Salminen PT, Laato MK. Surgical workload and cost of postoperative adhesion-related intestinal obstruction: importance of previous surgery. World J Surg. 2004;28:666-670.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  McCarthy JD. Further experience with the Childs-Phillips plication operation. Am J Surg. 1975;130:15-19.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Holland-Cunz S, Boelter AV, Waag KL. Protective fibrin-sealed plication of the small bowel in recurrent laparotomy. Pediatr Surg Int. 2003;19:540-543.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Sprouse LR 2nd, Arnold CI, Thow GB, Burns RP. Twelve-year experience with the Thow long intestinal tube: a means of preventing postoperative bowel obstruction. Am Surg. 2001;67:357-360.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Verco SJ, Peers EM, Brown CB, Rodgers KE, Roda N, diZerega G. Development of a novel glucose polymer solution (icodextrin) for adhesion prevention: pre-clinical studies. Hum Reprod. 2000;15:1764-1772.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Ivarsson ML, Falk P, Holmdahl L. Response of visceral peritoneum to abdominal surgery. Br J Surg. 2001;88:148-151.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Hill-West JL, Dunn RC, Hubbell JA. Local release of fibrinolytic agents for adhesion prevention. J Surg Res. 1995;59:759-763.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Falk K, Bjorquist P, Stromqvist M, Holmdahl L. Reduction of experimental adhesion formation by inhibition of plasminogen activator inhibitor type 1. Br J Surg. 2001;88:286-289.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Condon ET, Cahill RA, O'malley DB, Aherne NJ, Redmond HP. Evaluation of postoperative peritoneal adhesion formation following perioperative nicotine administration. J Surg Res. 2007;140:135-138.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Berkun Y, Ben-Chetrit E, Klar A, Ben-Chetrit E. Peritoneal adhesions and intestinal obstructions in patients with familial Mediterranean fever--are they more frequent? Semin Arthritis Rheum. 2007;36:316-321.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Cahill RA, Wang JH, Redmond HP. Enteric bacteria and their antigens may stimulate postoperative peritoneal adhesion formation. Surgery. 2007;141:403-410.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Petersen I. Antiangiogenesis, anti-VEGF(R) and outlook. Recent Results Cancer Res. 2007;176:189-199.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Kooloos WM, de Jong DJ, Huizinga TW, Guchelaar HJ. Potential role of pharmacogenetics in anti-TNF treatment of rheumatoid arthritis and Crohn's disease. Drug Discov Today. 2007;12:125-131.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Echtenacher B, Weigl K, Lehn N, Mannel DN. Tumor necrosis factor-dependent adhesions as a major protective mechanism early in septic peritonitis in mice. Infect Immun. 2001;69:3550-3555.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Wiczyk HP, Grow DR, Adams LA, O'Shea DL, Reece MT. Pelvic adhesions contain sex steroid receptors and produce angiogenesis growth factors. Fertil Steril. 1998;69:511-516.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Diamond MP, El-Hammady E, Munkarah A, Bieber EJ, Saed G. Modulation of the expression of vascular endothelial growth factor in human fibroblasts. Fertil Steril. 2005;83:405-409.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Howdieshell TR, Callaway D, Webb WL, Gaines MD, Procter CD Jr, Sathyanarayana , Pollock JS, Brock TL, McNeil PL. Antibody neutralization of vascular endothelial growth factor inhibits wound granulation tissue formation. J Surg Res. 2001;96:173-182.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Brown LF, Van de Water L, Harvey VS, Dvorak HF. Fibrinogen influx and accumulation of cross-linked fibrin in healing wounds and in tumor stroma. Am J Pathol. 1988;130:455-465.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Saltzman AK, Olson TA, Mohanraj D, Carson LF, Ramakrishnan S. Prevention of postoperative adhesions by an antibody to vascular permeability factor/vascular endothelial growth factor in a murine model. Am J Obstet Gynecol. 1996;174:1502-1506.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Grutzkau A, Kruger-Krasagakes S, Baumeister H, Schwarz C, Kogel H, Welker P, Lippert U, Henz BM, Moller A. Synthesis, storage, and release of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) by human mast cells: implications for the biological significance of VEGF206. Mol Biol Cell. 1998;9:875-884.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Boesiger J, Tsai M, Maurer M, Yamaguchi M, Brown LF, Claffey KP, Dvorak HF, Galli SJ. Mast cells can secrete vascular permeability factor/ vascular endothelial cell growth factor and exhibit enhanced release after immunoglobulin E-dependent upregulation of fc epsilon receptor I expression. J Exp Med. 1998;188:1135-1145.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Cahill RA, Wang JH, Soohkai S, Redmond HP. Mast cells facilitate local VEGF release as an early event in the pathogenesis of postoperative peritoneal adhesions. Surgery. 2006;140:108-112.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Grothey A. Antiangiogenic therapy in cancer: a new era has begun. Oncology (Williston Park). 2005;19:5-6.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Scappaticci FA, Fehrenbacher L, Cartwright T, Hainsworth JD, Heim W, Berlin J, Kabbinavar F, Novotny W, Sarkar S, Hurwitz H. Surgical wound healing complications in metastatic colorectal cancer patients treated with bevacizumab. J Surg Oncol. 2005;91:173-180.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Hamel KJ. Incidence of adhesions at repeat cesarean delivery. Am J Obstet Gynecol. 2007;196:e31-e32.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Bristow RE, Santillan A, Diaz-Montes TP, Gardner GJ, Giuntoli RL 2nd, Peeler ST. Prevention of adhesion formation after radical hysterectomy using a sodium hyaluronate-carboxymethylcellulose (HA-CMC) barrier: a cost-effectiveness analysis. Gynecol Oncol. 2007;104:739-746.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Tang CL, Jayne DG, Seow-Choen F, Ng YY, Eu KW, Mustapha N. A randomized controlled trial of 0.5% ferric hyaluronate gel (Intergel) in the prevention of adhesions following abdominal surgery. Ann Surg. 2006;243:449-455.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Tappenden P, Jones R, Paisley S, Carroll C. The cost-effectiveness of bevacizumab in the first-line treatment of metastatic colorectal cancer in England and Wales. Eur J Cancer. 2007;43:2487-2494.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Menzies D, Parker M, Hoare R, Knight A. Small bowel obstruction due to postoperative adhesions: treatment patterns and associated costs in 110 hospital admissions. Ann R Coll Surg Engl. 2001;83:40-46.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Tingstedt B, Isaksson J, Andersson R. Long-term follow-up and cost analysis following surgery for small bowel obstruction caused by intra-abdominal adhesions. Br J Surg. 2007;94:743-748.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Inoh K, Muramatsu H, Ochiai K, Torii S, Muramatsu T. Midkine, a heparin-binding cytokine, plays key roles in intraperitoneal adhesions. Biochem Biophys Res Commun. 2004;317:108-113.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Liu HJ, Wu CT, Duan HF, Wu B, Lu ZZ, Wang L. Adenoviral-mediated gene expression of hepatocyte growth factor prevents postoperative peritoneal adhesion in a rat model. Surgery. 2006;140:441-447.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Kosaka H, Yoshimoto T, Yoshimoto T, Fujimoto J, Nakanishi K. Interferon-gamma is a therapeutic target molecule for prevention of postoperative adhesion formation. Nat Med. 2008;14:437-441.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Hershlag A, Otterness IG, Bliven ML, Diamond MP, Polan ML. The effect of interleukin-1 on adhesion formation in the rat. Am J Obstet Gynecol. 1991;165:771-774.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Saba AA, Godziachvili V, Mavani AK, Silva YJ. Serum levels of interleukin 1 and tumor necrosis factor alpha correlate with peritoneal adhesion grades in humans after major abdominal surgery. Am Surg. 1998;64:734-736; discussion 737.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Cheong YC, Laird SM, Shelton JB, Ledger WL, Li TC, Cooke ID. The correlation of adhesions and peritoneal fluid cytokine concentrations: a pilot study. Hum Reprod. 2002;17:1039-1045.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Holschneider CH, Nejad F, Montz FJ. Immunomodulation with interleukin-10 and interleukin-4 compared with ketorolac tromethamine for prevention of postoperative adhesions in a murine model. Fertil Steril. 1999;71:67-73.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Montz FJ, Holschneider CH, Bozuk M, Gotlieb WH, Martinez-Maza O. Interleukin 10: ability to minimize postoperative intraperitoneal adhesion formation in a murine model. Fertil Steril. 1994;61:1136-1140.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Chegini N, Rong H, Bennett B, Stone IK. Peritoneal fluid cytokine and eicosanoid levels and their relation to the incidence of peritoneal adhesion. J Soc Gynecol Investig. 1999;6:153-157.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Saba AA, Kaidi AA, Godziachvili V, Dombi GW, Dawe EJ, Libcke JH, Silva YJ. Effects of interleukin-6 and its neutralizing antibodies on peritoneal adhesion formation and wound healing. Am Surg. 1996;62:569-572.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Ozgun H, Cevikel MH, Kozaci LD, Sakarya S. Lexipafant inhibits postsurgical adhesion formation. J Surg Res. 2002;103:141-145.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Reed KL, Fruin AB, Bishop-Bartolomei KK, Gower AC, Nicolaou M, Stucchi AF, Leeman SE, Becker JM. Neurokinin-1 receptor and substance P messenger RNA levels increase during intraabdominal adhesion formation. J Surg Res. 2002;108:165-172.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Gorvy DA, Herrick SE, Shah M, Ferguson MW. Experimental manipulation of transforming growth factor-beta isoforms significantly affects adhesion formation in a murine surgical model. Am J Pathol. 2005;167:1005-1019.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Diamond MP, El-Hammady E, Wang R, Saed G. Regulation of transforming growth factor-beta, type III collagen, and fibronectin by dichloroacetic acid in human fibroblasts from normal peritoneum and adhesions. Fertil Steril. 2003;79:1161-1167.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Freeman ML, Saed GM, Elhammady EF, Diamond MP. Expression of transforming growth factor beta isoform mRNA in injured peritoneum that healed with adhesions and without adhesions and in uninjured peritoneum. Fertil Steril. 2003;80 Suppl 2:708-713.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Okamoto Y, Takai S, Miyazaki M. Chymase inhibitor, BCEAB, suppressed peritoneal adhesion formation in hamster. J Surg Res. 2002;107:219-222.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Williams RS, Rossi AM, Chegini N, Schultz G. Effect of transforming growth factor beta on postoperative adhesion formation and intact peritoneum. J Surg Res. 1992;52:65-70.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Xu X, Rivkind A, Pappo O, Pikarsky A, Levi-Schaffer F. Role of mast cells and myofibroblasts in human peritoneal adhesion formation. Ann Surg. 2002;236:593-601.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Chegini N. The role of growth factors in peritoneal healing: transforming growth factor beta (TGF-beta). Eur J Surg Suppl. 1997;236:17-23.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Wasserberg N, Nunoo-Mensah JW, Ruiz P, Tzakis AG. The effect of immunosuppression on peritoneal adhesions formation after small bowel transplantation in rats. J Surg Res. 2007;141:294-298.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Okamoto Y, Takai S, Miyazaki M. Effect of chymase-dependent transforming growth factor beta on peritoneal adhesion formation in a rat model. Surg Today. 2004;34:865-867.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Saed GM, Kruger M, Diamond MP. Expression of transforming growth factor-beta and extracellular matrix by human peritoneal mesothelial cells and by fibroblasts from normal peritoneum and adhesions: effect of Tisseel. Wound Repair Regen. 2004;12:557-564.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Cheong YC, Shelton JB, Laird SM, Li TC, Ledger WL, Cooke ID. Peritoneal fluid concentrations of matrix metalloproteinase-9, tissue inhibitor of metalloproteinase-1, and transforming growth factor-beta in women with pelvic adhesions. Fertil Steril. 2003;79:1168-1175.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Ryan CK, Sax HC. Evaluation of a carboxymethylcellulose sponge for prevention of postoperative adhesions. Am J Surg. 1995;169:154-159; discussion 159-160.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Hobson KG, DeWing M, Ho HS, Wolfe BM, Cho K, Greenhalgh DG. Expression of transforming growth factor beta1 in patients with and without previous abdominal surgery. Arch Surg. 2003;138:1249-1252.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Krause TJ, Katz D, Wheeler CJ, Ebner S, McKinnon RD. Increased levels of surgical adhesions in TGFbeta1 heterozygous mice. J Invest Surg. 1999;12:31-38.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Holmdahl L, Kotseos K, Bergstrom M, Falk P, Ivarsson ML, Chegini N. Overproduction of transforming growth factor-beta1 (TGF-beta1) is associated with adhesion formation and peritoneal fibrinolytic impairment. Surgery. 2001;129:626-632.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Ghellai AM, Stucchi AF, Chegini N, Ma C, Andry CD, Kaseta JM, Burns JW, Skinner KC, Becker JM. Role of transforming growth factor beta-1 in peritonitis-induced adhesions. J Gastrointest Surg. 2000;4:316-323.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Bulbuller N, Ilhan YS, Kirkil C, Cetiner M, Gogebakan O, Ilhan N. Can angiotensin converting enzyme inhibitors prevent postoperative adhesions? J Surg Res. 2005;125:94-97.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Gago LA, Saed GM, Chauhan S, Elhammady EF, Diamond MP. Seprafilm (modified hyaluronic acid and carboxymethylcellulose) acts as a physical barrier. Fertil Steril. 2003;80:612-616.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Chegini N, Kotseos K, Zhao Y, Bennett B, McLean FW, Diamond MP, Holmdahl L, Burns J. Differential expression of TGF-beta1 and TGF-beta3 in serosal tissues of human intraperitoneal organs and peritoneal adhesions. Hum Reprod. 2001;16:1291-1300.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Yang XX, Shi HP, Hou LB. Chinese medicine compound Changtong oral liquid on postoperative intestinal adhesions. World J Gastroenterol. 2005;11:2967-2970.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Dwivedi AJ, Kuwajerwala NK, Silva YJ, Tennenberg SD. Effects of surgical gloves on postoperative peritoneal adhesions and cytokine expression in a rat model. Am J Surg. 2004;188:491-494.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Kurukahvecioglu O, Koksal H, Gulbahar O, Erdem O, Engin D, Yazicioglu O, Kerem M, Taneri F. Infliximab "TNF-alpha antagonist" decreases intraabdominal adhesions. Saudi Med J. 2007;28:1830-1835.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Kaidi AA, Gurchumelidze T, Nazzal M, Figert P, Vanterpool C, Silva Y. Tumor necrosis factor-alpha: a marker for peritoneal adhesion formation. J Surg Res. 1995;58:516-518.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Kaidi AA, Nazzal M, Gurchumelidze T, Ali MA, Dawe EJ, Silva YJ. Preoperative administration of antibodies against tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1) and their impact on peritoneal adhesion formation. Am Surg. 1995;61:569-572.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Ito T, Fraser IP, Yeo Y, Highley CB, Bellas E, Kohane DS. Anti-inflammatory function of an in situ cross-linkable conjugate hydrogel of hyaluronic acid and dexamethasone. Biomaterials. 2007;28:1778-1786.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Mirastschijski U, Johannesson K, Jeppsson B, Agren MS. Effect of a matrix metalloproteinase activity and TNF-alpha converting enzyme inhibitor on intra-abdominal adhesions. Eur Surg Res. 2005;37:68-75.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Ivarsson ML, Diamond MP, Falk P, Holmdahl L. Plasminogen activator/plasminogen activator inhibitor-1 and cytokine modulation by the PROACT System. Fertil Steril. 2003;79:987-992.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Rout UK, Oommen K, Diamond MP. Altered expressions of VEGF mRNA splice variants during progression of uterine-peritoneal adhesions in the rat. Am J Reprod Immunol. 2000;43:299-304.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Molinas CR, Campo R, Dewerchin M, Eriksson U, Carmeliet P, Koninckx PR. Role of vascular endothelial growth factor and placental growth factor in basal adhesion formation and in carbon dioxide pneumoperitoneum-enhanced adhesion formation after laparoscopic surgery in transgenic mice. Fertil Steril. 2003;80 Suppl 2:803-811.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Victory R, Saed GM, Diamond MP. Antiadhesion effects of docosahexaenoic acid on normal human peritoneal and adhesion fibroblasts. Fertil Steril. 2007;88:1657-1662.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Hoshino A, Kawamura YI, Yasuhara M, Toyama-Sorimachi N, Yamamoto K, Matsukawa A, Lira SA, Dohi T. Inhibition of CCL1-CCR8 interaction prevents aggregation of macrophages and development of peritoneal adhesions. J Immunol. 2007;178:5296-5304.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Holsti MA, Chitnis T, Panzo RJ, Bronson RT, Yagita H, Sayegh MH, Tzianabos AO. Regulation of postsurgical fibrosis by the programmed death-1 inhibitory pathway. J Immunol. 2004;172:5774-5781.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Mrstik M, Kotseos K, Ma C, Chegini N. Increased expression of interferon-inducible protein-10 during surgically induced peritoneal injury. Wound Repair Regen. 2003;11:120-126.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Berkkanoglu M, Zhang L, Ulukus M, Cakmak H, Kayisli UA, Kursun S, Arici A. Inhibition of chemokines prevents intraperitoneal adhesions in mice. Hum Reprod. 2005;20:3047-3052.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Zeyneloglu HB, Seli E, Senturk LM, Gutierrez LS, Olive DL, Arici A. The effect of monocyte chemotactic protein 1 in intraperitoneal adhesion formation in a mouse model. Am J Obstet Gynecol. 1998;179:438-443.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Gao Y, Luo L, He F. Effect of monocyte chemotactic protein-1 on the intraperitoneal adhesion formation. J Tongji Med Univ. 2000;20:340-342.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Zeyneloglu HB, Senturk LM, Seli E, Oral E, Olive DL, Arici A. The role of monocyte chemotactic protein-1 in intraperitoneal adhesion formation. Hum Reprod. 1998;13:1194-1199.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Chung DR, Chitnis T, Panzo RJ, Kasper DL, Sayegh MH, Tzianabos AO. CD4+ T cells regulate surgical and postinfectious adhesion formation. J Exp Med. 2002;195:1471-1478.  [PubMed]  [DOI]  [Cited in This Article: ]