• Non ci sono risultati.

IBD:Epidemiology and Risk Factors

N/A
N/A
Protected

Academic year: 2022

Condividi "IBD:Epidemiology and Risk Factors"

Copied!
8
0
0

Testo completo

(1)

of diseases affecting the bowel, the most common of which are ulcerative colitis and Crohn’s disease.

Whereas UC is characterised by a continuous distri- bution of mucosal/submucosal inflammation within the colon, CD may result in focal areas of disease in any part of the gastrointestinal tract from the mouth to the anus; the inflammation is transmural and almost inevitably progresses over time, often leading to structuring or fistulising complications. Extra- intestinal complications affecting eyes, skin and joints occur in both illnesses. Although inflammato- ry bowel disease is not common or highly fatal, it is important to public health because its highest inci- dence is early in life, its therapy involves major sur- gery including a curative colectomy for ulcerative colitis, and having the disease increases the risk of developing colon cancer.

There are literally hundreds of articles describing the incidence of ulcerative colitis and Crohn’s disease in many regions of the world. In general, the highest incidence rates and prevalence for both diseases have been reported in northern Europe [1–8], the United Kingdom [9–11], and North America [12–15], which

world such as southern or central Europe [16–18], Asia [19–22], Africa [23], and Latin America [24]

underscore the fact that the occurrence of IBD is a dynamic process. Incidence of UC, especially, is rising in several areas previously thought to have low inci- dence including Japan [21], South Korea [25], Singa- pore [22], northern India [26] and Latin America [24].

In most of these areas, however, CD remains rare.

In North America, incidence rates range from 2.2 to 14.3 cases/100 000 person-years for UC and from 3.1 to 14.6 cases/100 000 person-years for CD. Preva- lence ranges from 37 to 246 cases/100 000 persons for UC and from 26 to 199 cases/100 000 persons for CD [12–14].

The Multicenter European Collaborative Study on Inflammatory Bowel Disease (EC-IBD) reported blended incidence rates between 8.7 and 11.8 cases/100 000 person-years for UC and between 3.9 and 7.0 cases/100 000 person-years for CD [6]. The EC-IBD quantified the north–south gradient of inci- dence in Europe: the incidence rates for UC and for CD were respectively 40 and 80% higher in northern regions.

Fig. 1.UC incidence rates in Italy ([27]) Fig. 2.CD incidence rates in Italy ([27])

(2)

The last study from northern England [11] sug- gested that the prevalence of IBD in 1995 was 243/100 000 persons for UC and 144/100 000 persons for CD.

Italy, until some years ago, was considered among the countries with low incidence. In Italy, incidence rates are of 5.2 cases/100 000 person-years for UC and 2.3 cases/100 000 person-years for CD (Figs. 1, 2) [27].

The northern European studies based on popula- tion grounds have shown that there is no increase in overall mortality in IBD [28]. Only in CD has there been an increased mortality observed in the first few years after diagnosis in young patients. In some stud- ies, this was observed primarily in the females. The Italian studies data confirm other European studies [29–31].

In general, there is a slight female predominance in Crohn’s disease, especially among women in late adolescence and early adulthood, which suggests that hormonal factors may play a role in disease expres- sion. On the other hand, if there is a slight gender predominance regarding UC, it rests with males [15].

For UC, different patterns of incidence were observed for men and women aged 35 and over, with Fig. 4.Female annual incidence for UC ([32])

Fig. 5.Male annual incidence for UC ([32]

Fig. 6. Ratio of males to females according to smoking habits and age quartiles ([33])

Fig. 3.a Male and female incidence in different age groups for UC. b Male and female incidence in different age groups for CD ([6])

b a

(3)

increase with age. The correlation between the age groups and the M/F ratio could be explained by a greater tendency to smoke in men and therefore by a greater, gradual prevalence among them of giving up the habit in relation to, for example, the occurrence of cardiovascular problems (Fig. 6) [33].

Risk Factors

The etiology and pathogenesis of IBD probably involve an interaction between genetic and environ- mental factors; the precise mechanism for the begin- ning of the intestinal inflammation remains unclear.

There is increasing evidence that both environmental and host genetic factors are important in determining not only disease susceptibility, but also disease clinical course and response to therapy. Many factors have been suggested including family history of IBD, ciga- rette smoking, appendectomy, oral contraceptive agents, diet, breastfeeding, perinatal or early child- hood infections, hygienic factors and physical activity.

Evidence for genetic factors includes epidemiolog- ical data showing differences in IBD among different races and ethnic groups, familial aggregation and high concordance for the IBD type in monozygotic vs. dizygotic twins [34–35]. The incidence is highest among whites, somewhat lower among blacks and lowest among Asians, although it is increasing among the latter [36]. Among Caucasians, the most consistent observation has been that the Ashkenazi Jewish population has been shown to be at higher risk than other ethnic groups (ratio 4:1) and this risk is maintained irrespective of geographical location and time period [37].

A family history is the single greatest known risk factor for the development of IBD. Between 6 and 32% of patients with IBD have an affected first or sec- ond-degree relative and the prevalence of family his- tory is highest in Jewish patients and in patients with early onset of disease [38–39]. The risk is greatest among siblings, whereas it is lower among offsprings and second-degree relatives; the relative risk to a sib- ling of a patient with CD is 13–36% and for UC

in CD, but the discordance between monozygotic and dizygotic prevalence rates again argues for a genetic contribution to UC. However, genes alone are not sufficient to cause the disease and complex environ- mental triggers are required for disease expression [43–45].

In genetic terms, the IBD are “complex” because classical Mendelian inheritance attributable to a sin- gle gene locus is not exhibited. The model which appears most pertinent at the present time suggests that CD and UC may be related heterogeneous poly- genic disorders, sharing some but not all susceptibil- ity loci. The disease phenotype is likely to be deter- mined by the interaction between allelic variants at a number of loci and the interaction between genetic and environmental factors [46].

Recently the candidate genes have been selected on the basis of their location within an area of repli- cated linkage in multiplex IBD pedigrees (Fig. 7). The first susceptibility locus was identified in the pericen- tromeric region of chromosome 16 (IBD1) [47]. The importance of this locus has been confirmed by vari- ous groups, including the GISC [48] and the Interna- tional IBD Genetics Consortium [49]. In 2001, three independent studies reported the identification of the IBD1gene as NOD-2 [50–52]. Three coding region variants of this gene are associated with CD but not with UC. Patients carrying one high-risk allele have a 1.5–3-fold increased risk of developing CD, whereas those carrying two high-risk alleles have a 14–44-fold increased risk of developing CD [53]. The NOD2gene has recently been renamed CARD15(caspase recruit- ment domain gene) and is involved in the regulation of innate immune response and apoptosis; the leucine-rich repeat (LLR) domain of the gene appears to function as a sensor for bacterial products such as lipopolysaccharides (LPS; Fig. 8). Recently several genetic studies have been carried out in order to relate NOD2/CARD15variants to specific clinical fea- tures of patients. The studies have generally correlat- ed NOD2mutations with younger age at onset, ileal localisation and fibrostenotic behaviour of CD [54–57].

It has now been 23 years since the association of

(4)

non-smoking with UC was first identified by Harries et al. in 1982 [58], which was followed within 2 years by the observation that patients with CD were more often smokers [59]. This remarkable finding of

“opposite associations” for smoking with IBD has been the subject of intense scrutiny in the hope that it may help identify important pathogenic mecha- nisms responsible for the two conditions and per- haps provide the key to alternative therapeutic options.

In 1989, Calkins reviewed the available studies on the association with IBD. A meta-analysis showed an increased risk of ulcerative colitis among lifelong non-smokers and ex-smokers compared with cur- rent smokers (OR 1.64) [60]. The M/F ratio, at diag- nosis of UC, increases progressively with age in rela- tion to the ex-smoker’s habit compared with the M/F ratio that remains constant in non-smoking patients [33]. Moreover, it appears that ex-smokers are 70%

more likely than those who never smoked to develop UC. In contrast to UC, several studies have implicat- ed cigarette smoking as a risk factor for CD and the same meta-analysis suggests that in smokers the risk of having the disease is double in comparison to non- smokers. This association may not apply to all ethnic groups or geographic regions, a recent multicentre study performed in Israel has stressed the lack of association between smoking and CD and a negative association for UC [61]. Passive cigarette smoking has also been linked to IBD risk, but results have been conflicting. For UC the effect of passive smok- ing in childhood was comparable with that of active smoking in adulthood [62]. However, another study demonstrated that passive smoking exposure and maternal smoking at birth were significantly associ- ated with development of either subtype of IBD; in CD the association had a dose-response relationship [63].

Cigarette smoking may also influence the clinical

course of IBD. Active smokers were half as likely to be hospitalised for UC as non-smokers, whereas ex- smokers had higher hospitalisation and colectomy rates [64]. In another study, approximately 45% of UC patients who resumed smoking noted improve- ment in symptoms [65]. Moreover, significantly fewer smokers developed pouchitis compared with non-smokers among patients who had a restorative proctocolectomy for UC [66], and smoking is associ- ated with decreased risk of primary sclerosing cholangitis (PSC) [67].

Patients with CD who smoke (>10 cigarettes/day,

>150 cigarettes/year) are more likely to have ileal than colonic or ileocolonic involvement and fistulis- ing or stenosing than inflammatory disease [68-70].

Most of the studies show severe symptoms, more fre- quent relapses, higher recurrence rate after surgery and more frequent treatments with steroids and immunosuppressors in smokers compared with non- smokers [71-73]. Young women with CD who contin- ued to smoke, suffered more bowel and systemic symptoms in addition to more emotional dysfunc- tion than female non-smokers [74].

More recently the role of the appendix in IBD epi- demiology has become the subject of increasing interest. Appendectomy appears to be protective for the development of UC; important factors associated with a lower incidence of UC include an appendecto- my before the age of 20 and appendectomy for appendicitis or mesenteric lymphadenitis [75–77].

Similar to the effect of cigarette smoking, an appen- dectomy also influences the clinical course of UC. In fact, patients who developed UC after appendectomy were diagnosed at an older age, showed few recurrent symptoms and were significantly less likely to require colectomy [78–79].

On the other hand, many studies have suggested that appendectomy is associated with future risk of CD even after excluding a diagnosis within 1 year of Fig. 7.Significant areas of replicated linkage Fig. 8.Schematic representation of the NOD2/CARD15[51]

(5)

appendix in CD; it could be hypothesised that an etiopathogenic process may be phenotypically expressed in the form of appendicitis avoiding the expression of CD in the colon, similar to the protec- tive role of appendectomy regarding the develop- ment of UC. In addiction, the occurrence of an appendectomy before CD diagnosis seems to show a negative association with articular manifestations [81].

Several case-control and cohort studies have sug- gested an increased risk of IBD in women who take oral contraceptives. In some studies, a positive asso- ciation between oral contraceptives use and risk of CD was confined to women who were current smok- ers (OR 2.64) [62], whereas the opposite finding has been reported in other studies in which the elevated risk was found only among non-smokers (OR 3.1 vs.

0.91) [82]. There was no association between oral contraceptive use and UC (OR 0.70). Therefore, there is no evidence suggesting that women predisposed to the development of UC should be advised to avoid oral contraceptive use [83]. However, a 1995 meta- analysis demonstrated an elevated risk both for UC (OR 1.29) and CD (OR 1.44) [84]. Since this meta- analysis, other studies have been published with the same pattern of results: an overall weak association for both diseases but non-sufficiently compelling to infer a causal association [85-86]. The mechanism for this association is not definitively known, but it is thought that the thrombogenic properties of oral contraceptives, in the setting of a process of multifo- cal, microvascular gastrointestinal infarction, might play a role [87].

Because dietary agents are, next to bacterial anti- gens, the most common type of luminal antigen, it is logical to surmise that diet might play a role in the expression of IBD. Furthermore, differences in diet might explain the significant differences in IBD risk across geographic regions and the increase in IBD incidence in migrant populations. However, numer- ous studies have investigated dietary factors in IBD, and the methodological problems related to studies of this kind (type of population studied, recall bias, measure) have brought contradictive results. The

an inverse association between breastfeeding and IBD [86, 92, 93], in most cases the odds ratios were not statistically significant and other studies have not demonstrated such an association [94-95]. In gener- al, the association has been stronger for CD than for UC.

Perinatal infections, either in the infant or the mother, might influence the expression of IBD. It has been proposed that perinatal or childhood paramyx- oviral infection might result in persistent infection of the vascular endothelium of the mesentery, resulting in a chronic granulomatous vasculitis (CD) [96].

However, other studies have not been able to confirm these findings [97]. The same investigators who ini- tially proposed persistent measles infection as a cause of CD suggested that attenuated live measles virus vaccine might lead to IBD [98], but several case- control studies in different locales have not demon- strated significant differences in vaccination rates among IBD cases and unaffected controls [99]. Based on the available evidence, it would be difficult to con- clude that measles vaccination is a risk factor for IBD. A study of IBD from central Sweden noted that IBD patients with an infection or serious illness, mother or child, had a fourfold increased risk for IBD. In the same study, infants from families of low socioeconomic status were 3 times more likely later to develop IBD [100]. On the other hand, some have suggested that the absence of infections might be a risk for IBD; CD, but not UC, is more common in those who lived in houses with a hot water tap during childhood [101]. In addition, toothpaste has been proposed as a risk factor for CD; it contains such microparticles that could serve as a proxy measure of hygienic conditions or have an impact on the micro- biological intestinal flora [102]. However, no obser- vational studies have shown such an association implicating toothpaste as an independent risk factor.

Finally, Klein et al. have investigated the physical activity levels of patients with IBD and controls for the period prior to onset of the disease: IBD patients were over-represented in the low-activity group vs.

controls [103].

(6)

References

1. Langholz E, Munkholm P, Nielsen OH et al (1991) Incidence and prevalence of ulcerative colitis in Copenhagen county from 1962 to 1987. Scand J Gas- troenterol 26:1247–1256

2. Munkholm P, Langholz E, Nielsen OH et al (1992) Incidence and prevalence of Crohn’s disease in Copen- hagen county, 1962–1987: a sixfold increase incidence.

Scand J Gastroenterol 27:609–614

3. Gower-Rousseau C, Salomez JL, Marti R et al (1994) Incidence of inflammatory bowel disease in northern France. Gut 35:1433–1438

4. Moum B, Vatn MH, Ekbom A et al (1996) Incidence of Crohn’s disease in four counties in southeastern Nor- way, 1990–93: a prospective population study. Scand J Gastroenterol 31:355–361

5. Moum B, Vatn MH, Ekbom A et al (1996) Incidence of ulcerative colitis and indeterminate colitis in four counties of southeastern Norway, 1990–93: a prospective population study. Scand J Gastroenterol 31:362–366 6. Shivanada S, Lennard–Jones J, Logan R et al (1996)

Incidence of inflammatory bowel disease across Europe: Is there a difference between north and south?

Results of the European Collaborative Study on Inflammatory Bowel Disease (EC-IBD). Gut 39:690–697 7. Lapidus A, Bernell O, Hellers G et al (1997) Incidence of Crohn’s disease in Stockholm County 1955–1989.

Gut 41:480–486

8. Russel MG, Dorant E, Volovics A et al (1998) High incidence of inflammatory bowel disease in The Netherlands: a result of a prospective study. Dis Colon Rectum 41:33–40

9. Montgomery SM, Morris DL, Thompson NP et al (1998) Prevalence of inflammatory bowel disease in British 26 year olds: national longitudinal birth cohort.

BMJ 316:1058–1059

10. Yapp TR, Stenson R, Thomas GAO et al (2000) Crohn’s disease incidence in Cardiff from 1930: an update for 1991–1995. Eur J Gastroenterol Hepatol 12:907–911 11. Rubin GP, Hungin APS, Kelly PJ et al (2000) Inflam-

matory bowel disease: epidemiology and management in an English general practice population. Aliment Pharmacol Ther 14:1553–1559

12. Stowe SP, Redmond SR, Stormont JM et al (1990) An epidemiologic study of inflammatory bowel disease in Rochester, New York: hospital incidence. Gastroen- terology 98:104–110

13. Loftus EV Jr, Silverstein MD, Sandborn WJ et al (1998) Crohn’s disease in Olmsted County, Minnesota, 1940–1993: incidence, prevalence and survival. Gas- troenterology 114:1161–1168

14. Bernstein CN, Blanchard JF, Rawstorne P et al (1999) Epidemiology of Crohn’s disease and ulcerative colitis in a central Canadian province: a population-based study. Am J Epidemiol 149:916–924

15. Loftus EV Jr, Silverstein MD, Sandborn WJ et al (2000) Ulcerative colitis in Olmsted County, Minnesota, 1940–1993: incidence, prevalence and survival. Gut 46:336–343

16. Vucelic B, Korac B, Sentic M et al (1991) Epidemiolo- gy of Crohn’s disease in Zagreb, Yugoslavia: a ten-year prospective study. Int J Epidemiol 20:216–220

17. Vucelic B, Korac B, Sentic M et al (1991) Ulcerative colitis in Zagreb, Yugoslavia: incidence and prevalence 1980–1989. Int J Epidemiol 20:1043–1047

18. Mate–Jimenez J, Munoz S, Vicent D, Pajares JM (1994) Incidence and prevalence of ulcerative colitis and Crohn’s disease in urban and rural areas of Spain from 1981 to 1988. J Clin Gastroenterol 18:27–31

19. Fireman Z, Grossman A, Lilos P, Eshchar Y et al (1989) Epidemiology of Crohn’s disease in the Jewish popula- tion of central Israel, 1970–1980. Am J Gastroenterol 84:255–258

20. Grossman A, Fireman Z, Lilos P et al (1989) Epidemi- ology of ulcerative colitis in the Jewish population of central Israel, 1970–1980. Hepatogastroenterology 36:193–197

21. Morita N, Toki S, Hirohashi T et al (1995) Incidence and prevalence of inflammatory bowel disease in Japan: nationwide epidemiological survey during the year 1991. J Gastroenterol 30:1–4

22. Lee YM, Fock KM, See SJ et al (2000) Racial differences in the prevalence of ulcerative colitis and Crohn’s dis- ease in Singapore. J Gastroenterol Hepatol 15:622–625 23. Wright JP, Froggat J, O’Keefe EA et al (1986) The epi- demiology of inflammatory bowel disease in Cape Town 1980–1984. S Afr Med J 70:10–15

24. Linares de la Cal JA, Canton C, Hermida C et al (1999) Estimated incidence of inflammatory bowel disease in Argentina and Panama (1987–1993). Rev Esp Enferm Dig 91:277–286

25. Yang SK, Hong WS, Min YI et al (2000) Incidence and prevalence of ulcerative colitis in the Songpa-Kang- dong District, Seoul, Korea, 1986–1997. J Gastroen- terol Hepatol 15:1037–1042

26. Sood A, Midha V, Sood N et al (2003) Incidence and prevalence of ulcerative colitis in Punjab, North India.

Gut 52:1587–1590

27. Tragnone A, Corrao G, Miglio F et al (1996) Incidence of inflammatory bowel disease in Italy: a nationwide population-based study. Int J Epidemiol 25:1044–1052 28. Ekbom A, Helmick CG, Zack M et al (1992) Survival

and causes of death in patients with inflammatory bowel disease: a population-based study. Gastroen- terology 103:954–960

29. Palli D, Trallori G, Saieva C et al (1998) General and specific mortality of a population based cohort of patients with inflammatory bowel disease. Gut 42:175–179

30. Davoli M, Prantera G, Berto E et al (1997) Mortality among patients with ulcerative colitis: Rome 1970–1989. Eur J Epidemiol 13:189–194

31. Cottone M, Magliocco A, Rosselli M et al (1996) Mor- tality in patients with Crohn’s disease. Scand J Gas- troenterol 31:372–375

32. Binder V, Both H, Hansen PK et al (1982) Incidence and prevalence of ulcerative colitis and Crohn’s dis- ease in the county of Copenhagen, 1962–1978. Gas- troenterology 83:563–568

33. Riegler G, Tartaglione MT, Carratù R et al (2000) Age- related clinical severity at diagnosis in 1705 patients with ulcerative colitis: a study by GISC (Italian Colon- Rectum Study Group). Dig Dis Sci 45:462–465 34. Yang H, Rotter JI (2000) The genetic background of

inflammatory bowel disease. Hepatogastroenterology

(7)

39. Yang H, McElree C, Roth MP et al (1993) Familial empirical risks for inflammatory bowel disease: differ- ences between Jews and non-Jews. Gut 34:517–524 40. Orholm M, Munkolm P, Langholz E et al (1991) Famil-

ial occurrence of inflammatory bowel disease. N Engl J Med 324:84–88

41. Satsangi J, Rosenberg WMC, Jewell DP (1994) The prevalence of inflammatory bowel disease in relatives of patients with Crohn’s disease. Eur J Gastroenterol Hepatol 6:413–416

42. Peeters M, Nevens H, Baert F et al (1996) Familial aggregation in Crohn’s disease: increased age adjusted risk and concordance in clinical characteristics. Gas- troenterology 111:597–603

43. Tysk C, Lindberg E, Jarnerot G, Floderus-Myrhed B (1988) Ulcerative colitis and Crohn’s disease in an unselected population of monozygotic and dizygotic twins: a study of heritability and the influence of smoking. Gut 29:990–996

44. Orholm M, Binder V, Sorensen T, Kyvik K (1996) Inflammatory bowel disease in a Danish twin register.

Gut 39:A187

45. Thompson NP, Driscoll R, Pounder RE, Wakefield AJ (1996) Genetics versus environment in inflammatory bowel disease: results of a British twin study. BMJ 312:95–96

46. Satsangi J, Parkes M, Jewell DP (1997) The genetics of inflammatory bowel disease: recent progress. In:

McLeod RS, Martin F, Sutherland LR et al (eds) Inflammatory bowel diseases. Kluwer, Lancaster, pp 21–28

47. Hugot JP, Laurent-Puig P, Gower-Rousseau C et al (1996) Mapping of a susceptibility locus for Crohn’s disease on chromosome 16. Nature 379:821–823 48. Annese V, Latiano A, Bovio P et al (1999) Genetic

analysis in Italian families with inflammatory bowel disease supports linkage to the IBD1 locus: a GISC study. Eur J Hum Genet 7:567–573

49. The IBD International Genetics Consortium (2001) International collaboration provides convincing link- age replication in complex disease through analysis of a large pooled data set: Crohn’s disease and chromo- some 16. Am J Hum Genet 68:1165–1171

50. Hugot JP, Chamaillard M, Zouali H et al (2001) Asso- ciation of NOD2 leucine-rich repeat variants with sus- ceptibility to Crohn’s disease. Nature 411:599–603 51. Ogura Y, Bonen DK, Inohara N et al (2001) A

frameshift mutation in NOD2 associated with suscep- tibility to Crohn’s disease. Nature 411:603–606

ated with fistulizing and fibrostenotic phenotypes in Crohn’s disease. Gastroenterology 122:2091–2092 56. Abreu MT, Taylor KD, Lin YC et al (2002) Mutations

in NOD2 are associated with fibrostenosing disease in patients with Crohn’s disease. Gastroenterology 123:679–688

57. Louis E, Hugot JP, Colombel JF et al (2002) NOD2/CARD15 mutations in Crohn’s disease are associated with young age at diagnosis and ileal loca- tion. Gastroenterology 122:M1405

58. Harries AD, Baird A, Rhodes J (1982) Non-smoking: a feature of ulcerative colitis. BMJ 284:706

59. Sommerville KW, Logan RF, Edmond M, Langman MJ (1984) Smoking and Crohn’s disease. BMJ 289:954–956 60. Calkins BM (1989) A meta-analysis of the role of smoking in inflammatory bowel disease. Dig Dis Sci 34:1841–1854

61. Reif S, Lavy A, Keter D et al (2000) Lack of association between smoking and Crohn’s disease but the usual association between ulcerative colitis in Jewish patients in Israel: a multicenter study. Am J Gastroen- terol 95:474–478

62. Sandler RS, Sandler DP, McDonnel CW, Wurzelmann JI (1992) Childhood exposure to environmental tobac- co smoke and the risk of ulcerative colitis. Am J Epi- demiol 135:603–608

63. Lashner BA, Shaheen NJ, Hanauer SB, Kirschner BS (1993) Passive smoking is associated with an increased risk of developing inflammatory bowel disease in chil- dren. Am J Gastroenterol 88:1093–1099

64. Boyko EJ, Perera DR, Koepsell TD et al (1988) Effects of cigarette smoking on the clinical course of ulcera- tive colitis. Scand J Gastroenterol 23:1147–1152 65. Rudra T, Motley R, Rhodes J (1989) Does smoking

improve colitis? Scand J Gastroenterol Suppl 170:61–63

66. Merret MN, Mortensen N, Kettlewell M, Jewell DO (1996) Smoking may prevent pouchitis in patients with restorative proctocolectomy for ulcerative colitis.

Gut 38:362–364

67. van Erpecum KJ, Smits SJ, van de Meeberg PC et al (1996) Risk of primary sclerosing cholangitis is associ- ated with non-smoking behaviour. Gastroenterology 110:1503–1506

68. Lindberg E, Jarnerot G, Huitfeldt B (1992) Smoking in Crohn’s disease: effect on localisation and clinical course. Gut 33:779–782

69. Russel MG, Volovics A, Schoon EJ et al (1998) Inflam- matory bowel disease: Is there any relation between

(8)

smoking status and disease presentation? Inflamm Bowel Dis 4:182–186

70. Picco MF, Bayless TM (2003) Tobacco consumption and disease duration are associated with fistulizing and stricturing behaviours in the first 8 years of Crohn’s disease. Am J Gastroenterol 98:363–368 71. Sutherland LR, Ramcharan S, Bryant H, Fick G (1990)

Effect of cigarette smoking on recurrence of Crohn’s disease. Gastroenterology 98:1123–1128

72. Cottone M, Rosselli M, Orlando A et al (1994) Smok- ing habits and recurrence in Crohn’s disease. Gas- troenterology 106:643–648

73. Cosnes J, Carbonnel F, Carrat F et al (1999) Effects of current and former cigarette smoking on the clinical course of Crohn’s disease. Aliment Pharmacol Ther 13:1403–1411

74. Russel MG, Nieman FH, Bergers JM, Stockbrugger RW (1996) Cigarette smoking and quality of life in patients with inflammatory bowel disease. Eur J Gastroenterol Hepatol 8:1075–1081

75. Rutgeerts P, D’Haens G, Hiele M et al (1994) Appen- dectomy protects against ulcerative colitis. Gastroen- terology 106:1251–1253

76. Parrello T, Pavia M, Angelillo G et al (1997) Appen- dicectomy is an independent protective factor for ulcerative colitis: results of a multicenter case control study. Ital J Gastroenterol Hepatol 29:208–213 77. Andersson RE, Olaison G, Tysk C, Ekbom A (2001)

Appendectomy and protection against ulcerative coli- tis. N Engl J Med 344:808–814

78. Naganuma M, Iizuka B, Torii A et al (2001) Appendec- tomy protects against the development of ulcerative colitis and reduces its recurrence: results of a multi- center case-controlled study in Japan. Am J Gastroen- terol 96:1123–1126

79. Cosnes J, Carbonnel F, Beaugerie L et al (2002) Effects of appendicectomy on the course of ulcerative colitis.

Gut 51:803–807

80. Andersson RE, Olaison G, Tysk C, Ekbom A (2003) Appendectomy is followed by increased risk of Crohn’s disease. Gastroenterology 124:40–46

81. Riegler G, Caserta L, Esposito I et al (2005) Worse clin- ical course of disease in Crohn’s patients with previous appendectomy. Eur J Gastroenterol Hepatol 17:623–627 82. Katschinski B, Fingerle D, Scherbaum B, Goebell H (1993) Oral contraceptive use and cigarette smoking in Crohn’s disease. Dig Dis Sci 38:1596–1600

83. Lashner BA, Kane SV, Hanauer SB (1990) Lack of asso- ciation between oral contraceptive use and ulcerative colitis. Gastroenterology 99:1032–1036

84. Godet PG, May GR, Sutherland LR (1995) Meta-analy- sis of the role of oral contraceptive agents in inflam- matory bowel disease. Gut 37:668–673

85. Boyko EJ, Theis MK, Vaughan TL, Nicol-Blades B (1994) Increased risk of inflammatory bowel disease associated with oral contraceptive use. Am J Epidemi- ol 140:268–278

86. Corrao G, Tragnone A, Caprilli R et al (1998) Risk of inflammatory bowel disease attributable to smoking,

oral contraception and breastfeeding in Italy: a nation- wide case-control study. Int J Epidemiol 27:397–404 87. Wakefield AJ, Sawyerr AM, Hudson M et al (1991)

Smoking, the oral contraceptive pill and Crohn’s dis- ease. Dig Dis Sci 36:1147–1150

88. Riordan AM, Ruxton CH, Hunter JO (1998) A review of associations between Crohn’s disease and con- sumption of sugars. Eur J Clin Nutr 52:229–338 89. Russel MG, Engels LG, Muris JW et al (1998) “Modern

life” in the epidemiology of inflammatory bowel dis- ease: a case-control study with special emphasis on nutritional factors. Eur J Gastroenterol Hepatol 10:243–249

90. Reif S, Klein I, Lubin F et al (1997) Pre-illness dietary factors in inflammatory bowel disease. Gut 40:754–760 91. Persson PG, Ahlbom A, Hellers G (1992) Diet and inflammatory bowel disease: a case-control study. Epi- demiology 3:47–52

92. Koletzko S, Sherman P, Corey M et al (1989) Role of infant feeding practices in development of Crohn’s disease in childhood. BMJ 298:1617–1618

93. Rigas A, Rigas B, Glassman M et al (1993) Breastfeed- ing and maternal smoking in the etiology of Crohn’s disease and ulcerative colitis in childhood. Ann Epi- demiol 3:387–392

94. Thompson NP, Montgomery SM, Wadsworth ME et al (2000) Early determinants of inflammatory bowel dis- ease: use of two national longitudinal birth cohorts.

Eur J Gastroenterol Hepatol 12:25–30

95. Ekbom A, Adami HO, Helmick CG et al (1990) Perina- tal risk factors for inflammatory bowel disease: a case- control study. Am J Epidemiol 132:1111–1119 96. Thompson NP, Pounder RE, Wakefield AJ (1995) Peri-

natal and childhood risk factors for inflammatory bowel disease: a case-control study. Eur J Gastroen- terol Hepatol 7:385–390

97. Wakefield AJ, Ekbom A, Dhillon AP et al (1995) Crohn’s disease: pathogenesis and persistent measles virus infection. Gastroenterology 108:911–916 98. Pardi DS, Tremaine WJ, Sandborn WJ et al (1999)

Perinatal exposure to measles virus is not associated with the development of inflammatory bowel disease.

Inflamm Bowel Dis 5:104–106

99. Thompson NP, Montgomery SM, Pounder RE, Wake- field AJ (1995) Is measles vaccination a risk factor for inflammatory bowel disease? Lancet 345:1071–1074 100. Davis RL, Kramarz P, Bohlke K et al (2001) Measles-

mumps-rubella and other measles-containing vac- cines do not increase the risk for inflammatory bowel disease: a case-control study from the Vaccine Safety Datalink Project. Arch Pediatr Adolesc Med 155:354–359 101. Gent AE, Hellier MD, Grace RH et al (1994) Inflamma- tory bowel disease and domestic hygiene in infancy.

Lancet 343:766–767

102. Sullivan SN (1990) hypothesis revisited: toothpaste and the cause of Crohn’s disease. Lancet 336:1096–1097 103. Klein I, Reif S, Farbstein H et al (1998) Preillness non dietary factors and habits in inflammatory bowel dis- ease. Ital J Gastroenterol Hepatol 30:247–251

Riferimenti

Documenti correlati

Late-life depression and risk of vascular dementia and Alzheimer’s disease: Systematic review and meta-analysis of community-based cohort studies [Internet]. 202, British Journal

where Attacks c ,t is a dummy for having at least one terrorist attack in country c and month t; Organized crime c is the presence of criminal organizations in country c;

I l caso da noi riferito, pur inquadrato nell'ambito della iperplasia angiofollicolare (tipo ialino-vascolare) si è pre- sentato sotto una forma alquanto atipica per la

Spazi e immaginari sono dei composti, dei prodotti culturali pieni (densi) di narrazioni, discorsi, esteticità?, ma quando li guardiamo siamo presi dal mondo della finzione, dal

At the present it’s not possible to discriminate which patients, among those with diabetes insipidus, may have a higher risk to have neurodegenerative lesions and, most of all,

This double-blind, multicenter, randomized study compared melphalan–predni- sone–lenalidomide induction followed by lenalidomide maintenance (MPR-R) with

The aim of this study was to assess the mineral composition, antioxidant activity, total phenolics and tar- get polyphenols of two-seed propagated artichoke cultivars ‘Romolo’