Maintenance of Remission in Ulcerative Colitis


1. Disease-related

   (a) Extent of disease

   (b) Activity and severity of disease

    • Frequency of flares

    • Failure of prior maintenance therapy

    • Severity of the most recent flare

    • Mode of treatment of the most recent flare

2. Patient-related

   (a) Patient preferences

   (b) Adherence

3. Medication-related

   (a) Effectiveness

   (b) Appropriateness

   (c) Safety

   (d) Cost

   (e) Availability


Adapted from [3, 4]




Table 39.2
Risk factors for relapse in ulcerative colitis













































 1. Adherence to treatment

 2. Young, single status

 3. Low-fibre diet

 4. Intestinal infections

 5. Medications

    (a) Nonsteroidal anti-inflammatory drugs (NSAIDs)

    (b) Antibiotics

 6. Seasonal factors

 7. Stressful events of life

 8. Clinical factors

    (a) Disease activity and severity

    • Shorter interval between relapses

    • Severity of the most recent relapse

    • Need for steroids

    • Slow response to steroids

    (b) Active extra-intestinal manifestations

 9. Persistent elevation of serum C-reacting protein

10. Endoscopic and histologic factors

    (a) Lack of mucosal healing

    (b) Persistent basal plasmacytosis and active inflammatory infiltrate at colonic histology


Adapted from [3, 4, 15, 16, 18]



Table 39.3
Demographic, environmental, clinical, serological, endoscopic and histologic risk factors for relapse of ulcerative colitis

















































1. Demographic

   (a) Young age

   (b) Being single

2. Environmental

   (a) Perceived and/or actual “stressful” events of life

   (b) Low-fibre diet

   (c) Infections

   (d) Nonsteroidal anti-inflammatory drugs

   (e) Antibiotics

3. Clinical

   (a) Shorter interval between relapses

   (b) Severity of the most recent relapse

    • Need for steroids

    • Slow response to steroids

   (c) Presence of active extra-intestinal manifestations

4. Serological

   (a) Persistent elevation of serum C-reacting protein

5. Endoscopic

   (a) Lack of mucosal healing

6. Histology at colonic (rectal) biopsies

   (a) Persistent basal plasmacytosis

   (b) Active inflammatory infiltrate despite clinical remission


Extent of disease is important. Patients with extensive colitis (E-UC) are at higher risk for complications and colectomy and usually need more intensive therapy than patients with shorter extent of disease. In contrast, patients with distal disease are in higher needs of topical therapy. Suboptimal treatment may lead onto proximal extension of ulcerative proctitis (UP) or left-sided colitis (L-UC) in up to 50 % of patients and usually marks a more refractory course of disease [17, 18]. Maintenance therapy should adapt to the clinical pattern and course of disease. A severe flare or frequent relapses indicate failure of prior maintenance therapy and mandate intensification of treatment to recapture steroid-free remission.

Adherence is the most important factor that determines relapse. Non-adherence to mesalamine increases the likelihood of relapse at least five times [19, 20].

Finally, close monitoring is vital to maintain sustained remission even in asymptomatic patients in order to prevent the long-term sequelae of disease.



Effectiveness of Medications for Maintenance of Remission



Aminosalicylates



Sulfasalazine and Oral Aminosalicylates


Several meta-analyses have assessed the efficacy and safety profile of sulfasalazine and the newer aminosalicylates for remission maintenance of quiescent UC [2125]. Sulfasalazine in daily doses of 1–4 g has demonstrated a dose–response effect and can maintain remission in 71–89 % of patients [26, 27]. In the 2006 Cochrane systematic review, oral 5-ASA preparations at doses ranging from 0.8 to 4 g per day were superior to placebo in maintaining clinical and/or endoscopic remission with a pooled odds ratio of 0.47 [95 % confidence interval (CI), 0.36–0.62 and a number needed to treat (NNT) of 6] [20]. This meta-analysis demonstrated that sulfasalazine was more effective [OR 1.29 (95 % CI, 1.05–1.57)] and had a similar adverse event profile to mesalamine [OR 1.16 and 1.31, respectively] but has been criticised for including only patients who tolerated sulfasalazine. These results have been challenged by the recent large meta-analysis of Ford et al. [22] that confirmed that sulfasalazine, mesalamine, balsalazide and olsalazine were effective at preventing clinical relapse [11 trials of 6–12 months duration, 849 5-ASA treated vs. 653 placebo-treated patients, RR of 0.65, p = 0.02, NNT of 4], endoscopic relapse (3 trials, RR of 0.56, p = 0.01, NNT of 4) or both (6 trials, RR of 0.59, NNT of 4) without any significant differences between 5-ASA formulations. Nonetheless, subgroup analysis showed superiority over placebo of sulfasalazine (RR of 0.45, NNT of 3) and mesalamine (RR of 0.65, NNT of 4) but not olsalazine (RR of 0.72). In another two meta-analyses, sulfasalazine and mesalamine were equally effective to balsalazide for remission maintenance, but balsalazide was superior to sulfasalazine for withdrawals due to adverse events (RR of 0.17, p = 0.001) [23, 24]. Regarding comparisons between mesalamine formulations, Ito et al. [25] performed a 12-month non-inferiority trial and found no significant differences in the proportion of patients without bloody stools between a 2.4 g/day pH-dependent release and a 2.25 g/day time-dependent mesalamine formulation. 5-ASA formulations were shown to maintain remission irrespective of the extent of disease [26, 27].

Based on these results, sulfasalazine should be recommended as the first-choice maintenance treatment in UC due to its efficacy and lower cost, whereas mesalamine, olsalazine and balsalazide should be reserved for patients intolerant of sulfasalazine [4]. However, as the optimal maintenance dose of sulfasalazine (4 g/day) cannot be tolerated by a significant proportion of patients, most gastroenterologists incline to use the newer 5-ASA preparations because they are better tolerated at greater than equivalent doses of sulfasalazine [3, 28, 29].

The optimal maintenance mesalamine dose is unknown. An Italian study documented that 1.2 g/day oral mesalamine was equally effective to 2.4 g/day for remission maintenance [12]. In another study, the minimum effective dose of oral mesalamine was 0.8 g/day, but an incremental benefit for patients receiving 1.6 g/day was shown [30]. However, although a clear dose–response at doses over 0.8 g 5-ASA has never been convincingly demonstrated, dose-ranging studies in patients who required doses of mesalamine up to 4.8 g/day to achieve remission have not been performed [31]. In fact, there is some evidence that higher doses may be more effective. Thus, in a 1-year prospective controlled trial, per protocol analysis of data demonstrated that 3.0 g of oral mesalamine (Salofalk®) once daily was superior to 1.5 g once daily or 0.5 g three times weekly (86 %, 67 % and 78 %, respectively, p = 0.024) [32]. In a meta-analysis of seven randomised clinical trials, 5-ASA doses greater than 2.0 g/day were more likely to reduce the risk for relapse for 6–12 months compared to doses lower than 2.0 g/day, but the quality of the trials that were subjected to analysis was not optimal [22]. Indirect evidence comes also from a post hoc analysis of two clinical trials with Multi-Matrix (MMX) mesalamine in mild-to-moderate UC [10] where 196 of 218 (89.9 %) patients who achieved remission and were maintained on the induction dose of mesalamine were still relapse free after 1 year, indicating that the higher doses that induced remission are expected to maintain also high rates of remission.

Once-daily dose of mesalamine is equally effective and safe, better tolerated and preferred by patients over divided daily doses [12, 13, 3335]. This effect appears to be similar across all studies irrespective of the mesalamine formulation. A single tablet of 1.2 g MMX mesalamine was as effective as twice-daily dosing (2.4 g) in maintaining clinical remission (88.9 % vs. 93.2 %) and combined clinical and endoscopic remission (64.4 % vs. 68.5 %) for 12 months; both dosing regimens demonstrated a similar adverse event profile and very high adherence rates [13]. In another single-blinded study, 2 g delayed-release 5-ASA granules (Pentasa®) once daily were superior to 1 g twice daily in maintaining remission of quiescent UC for 1 year (70.9 % vs. 58.9 %, respectively, p = 0.024) [33]. In a very large study, 1.6–2.4 g mesalamine (Asacol®) once daily was not inferior to twice-daily dosing in maintaining remission of quiescent UC for 1 year (85.4 % vs. 85.4 %, respectively) [34]. Additionally, mesalamine granules once daily at a dose of 1.5 g daily was more effective than placebo to maintain remission over a 6-month period. Finally, 2.4 g MMX mesalamine once daily was equally effective to 2.4 g/day pH-modified release mesalamine (Asacol®) in divided doses at preventing clinical and/or endoscopic relapse of quiescent UC [35].


Topical 5-ASA


The efficacy and safety of various formulations (suppositories, foam and liquid or gel enemas) and dosing regimens of rectally administered mesalamine have been assessed in controlled clinical trials and case series for maintenance of remission of UP, “distal” colitis (“proctosigmoiditis”) and L-UC [3641]. 5-ASA is delivered to the upper rectum by suppositories and to the rectosigmoid area by foam enemas. Liquid enemas may deliver the active compound up to the splenic flexure, but the actual area of distribution varies between individuals, depending on the length of the sigmoid colon, the volume and the viscosity of the enema.

The advantage of the topical (rectal) therapy is that delivering the active compound directly to the affected area in less frequent dosing schedules increases its effectiveness and reduces systemic availability and adverse effects. In controlled clinical trials, the overall efficacy of topical mesalamine to prevent clinical and/or endoscopic relapse of UP and L-UC for 1 year ranges between 52 and 80 % [4]. Corresponding figures for placebo-treated patients range between 11 % and 53 % and are statistically lower than active treatment in almost all clinical trials. Topical mesalamine administered intermittently three times or even twice weekly is at least equally effective to oral sulfasalazine and mesalamine [18, 38, 42, 43]. However, unlike older mesalamine formulations, oral MMX mesalamine which aims at delivering 5-ASA more evenly throughout the colon has not been tested against topical therapy in quiescent UC. The frequency of topical administration depends largely on patient tolerance, frequency of prior flares and treatment regimen used to induce remission of the most recent flare and may range from 1 g per day to 1–4 g every third day [18, 44]. Topical steroids including the newer formulations with low systemic bioavailability have not shown maintenance efficacy [3, 4, 18, 45].


Combination of Oral and Topical 5-ASA


The combination of oral and topical 5-ASA is superior to oral or topical therapy alone at preventing relapse of quiescent UC [46, 47]. There are no additional safety signals compared to oral or topical therapy, but the long-term tolerance of treatment is debatable. Topical therapy is usually administered intermittently. Combined treatment should be considered as an escalation of therapy for patients who have relapsed despite optimal oral or topical 5-ASA monotherapy [3, 4, 18, 44].


Thiopurines


The quality of evidence for the efficacy of thiopurines, azathioprine (AZA) and 6-mercaptopurine (6-MP) in maintaining remission of UC is rather poor. It comes from retrospective uncontrolled observational cohorts from tertiary centres, uncontrolled case series and small prospective controlled trials [4863]. These studies being few, heterogeneous, of limited size, diverse methodology and varied outcome measures have yielded conflicting results. Consequently, meta-analyses and systematic reviews of the literature cannot offer a precise estimate of the efficacy of thiopurines as maintenance agents in UC. Thus, although in the 2007 Cochrane meta-analysis [64] based on four randomised controlled trials of 12-month duration AZA was superior to placebo at preventing relapse with an odds ratio of 0.41 (95 % CI 0.24–0.70), Leung et al. [65] in their recent review suggested that AZA is only modestly effective in inducing and maintaining remission in UC. In another meta-analysis of six trials that included 124 patients treated with AZA or 6-MP, the mean maintenance efficacy of thiopurines was 60 % versus only 37 % in controls (OR 2.56, 95 % CI 1.51–4.34) [66]. The pooled OR was 2.59 (95 % CI 1.26–5.3) with an absolute risk reduction of 23 % and a NNT of 5 when only studies of thiopurines versus placebo were analysed. However, these results have been criticised for methodological flaws of the studies that were included in the meta-analysis. Khan et al. [67] analysed recently only three randomized controlled trials, all with “unclear risk of bias”, that included 127 patients with quiescent UC followed for 9–12 months and found that AZA was superior to placebo at preventing relapse (RR = 0.60; 95 % CI = 0.37–0.95; p = 0.03, NNT = 4). Additionally another meta-analysis found similar findings.

Despite published evidence, thiopurines have been recommended by experts and societies’ practice guidelines [3, 4, 18] for nearly 40 years as steroid-sparing and remission maintenance agents (see Table 39.4). Thiopurines are especially indicated for patients who cannot tolerate, respond, be waned from or relapse early after discontinuation of oral steroids, for thiopurine-naïve patients who have achieved long-term sustained remission on infliximab (IFX) monotherapy or combined with AZA and are considered for discontinuation of IFX and, finally, for hospitalised thiopurine-naïve patients responding to intravenous steroids or to second-line therapy with a fast-acting agent, such as intravenous cyclosporine or tacrolimus who need to be bridged to a steroid-sparing agent [3, 68, 69]. For these indications, thiopurines are likely to maintain clinical remission in approximately 40–70 % of patients’ mucosal healing [4] and steroid sparing [70], resulting in a significant reduction in hospitalisations and colectomies [58].


Table 39.4
Indications for thiopurines as maintenance agents in ulcerative colitis























1. Intolerance of 5-ASA

2. Frequent relapses despite maximum dose of oral and topical 5-ASA

3. Intolerance of steroids

4. Mild-to-moderate steroid-dependent disease

5. Relapse soon after discontinuation of oral steroids

6. Oral steroid-refractory disease in combination with infliximab

7. Clinical response/remissiona achieved on

  (a) IV steroids

  (b) IV cyclosporine or IV tacrolimus, infliximab, adalimumab, golimumab or vedolizumab


aThiopurine-naïve patients

The daily dose of AZA is 2.5 mg/kg and of 6-MP is 1–1.5 mg/kg. Treatment may be started at low doses (50 mg AZA or 25 mg 6-MP) and increased gradually if tolerated by 50 mg for AZA or 25 mg for 6-MP in order to achieve the target dose over a period of 2 months. A dosing strategy based on TPMT testing is discussed in Chaps. 13 and 14. Frequent white blood cell (WBC) and platelet count should be performed to avoid early leucopenia. Since late leucopenia and liver toxicity may develop at any time, patients should be monitored at regular intervals with WBC and platelet counts and liver function tests. If the WBC and/or the platelet count drops below 3.000/ml and 80.000/ml, respectively, thiopurines should be tapered or stopped temporarily. Adherence can be monitored by frequent consultations, checking the mean volume of red blood cells (MCV) or by measuring 6-MP metabolites. There is insufficient evidence to recommend monitoring the response to thiopurines by sequential measurements of 6-MP metabolites over the traditional approach of frequent clinical consultations and laboratory tests [4].

Relative leucopenia, a higher MCV and being older may predict response to AZA [54]. Efficacy appears to be higher in patients with shorter disease duration [70, 71]. There are no studies comparing the efficacy of AZA to 6-MP for remission maintenance. The doses of 6-MP in observational cohorts are probably lower to AZA doses, but the overall efficacy appears to be similar [4, 4954].

Co-administration of thiopurines and 5-ASA increases 6-TGN metabolites in a dose-dependent manner. This interaction may result in myelotoxicity in a small proportion of patients but may theoretically be beneficial for patients who are refractory to thiopurines [72]. Whether these are clinically meaningful is questionable. Two small studies and a Cochrane meta-analysis have suggested that 5-ASA offers no advantage to AZA for the maintenance of remission [63, 73, 74]. However, many gastroenterologists incline to continue 5-ASA as a colorectal cancer preventive agent.

Although there is limited evidence to recommend certain duration of treatment [3], treatment should probably be indefinite [75]. AZA-withdrawal studies have demonstrated consistently that UC will inevitably relapse shortly after abrupt cessation of treatment [63, 72]. In an Oxford cohort, the proportion of IBD patients in remission after withdrawal of AZA was 0.63 at 1 year and 0.35 at 5 years [54]. It is also unclear whether disease extent, duration of treatment and concomitant use of 5-ASA influence relapse [54, 7274]. Prolonged use of thiopurines may increase the risk of lymphoma, but the magnitude of this risk has not been completely defined [3, 4, 76, 77]. A recent meta-analysis suggests that a minimum of 1 year is needed for the risk of lymphoma and also that this occurs more commonly in men than women. Men have a greater risk than women (RR = 2.05; p < 0.05); both sexes were at increased risk for lymphoma (SIR for men = 3.60; 95 % CI, 2.68–4.83 and SIR for women = 1.76, 95 % CI, 1.08–2.87). Patients younger than 30 years had the highest RR (SIR = 6.99; CI, 2.99–16.4); younger men had the highest risk. The absolute risk was highest in patients older than 50 years (1:377 cases per patient year). In any case the risk of prolonged treatment needs to be balanced against colectomy at an individual level.

It is also questionable whether the full dose of thiopurines is needed to maintain long-term remission. However, underdosing may lead onto relapse of disease without preventing drug toxicity.


Anti-TNFα Agents



Infliximab


Two large studies, ACT 1 and ACT 2, have assessed the efficacy of IFX in patients with moderate-to-severe UC refractory to steroids, immunomodulators (ACT 1 and 2) and/or 5-ASA (ACT 2) [7]. Remission rates at week 30 (ACT 1 and 2) and at week 54 (ACT 1) were assessed only in patients who had achieved clinical response or remission at week 8 after induction with IFX (5 or 10 mg/kg at weeks 0, 2 and 6) or placebo and continued IFX schedule therapy (5 or 10 mg/kg) or placebo every 8 weeks. Remission rates were significantly higher for IFX groups in ACT 2 at week 30 (26 % vs. 36 % vs. 11 % for 5 mg/kg and 10 mg/kg IFX or placebo, respectively) and in ACT 1 at week 54 (34 % for combined IFX groups vs. 17 % for placebo). In ACT 1, complete mucosal healing defined as a Mayo score 0 or 1 at week 54 was superior for IFX than placebo-treated patients (55 %, 57 % and 22 %, for 5 mg/kg, 10 mg/kg IFX and placebo, respectively). Although the gain in steroid-free remission at week 54 (ACT-1) was only 14 % for the 5 mg/kg arm and 9 % for the 10 mg/kg IFX over placebo (10 %), IFX resulted in a significant reduction in hospitalisations and colectomies [78]. The degree of mucosal healing at week 8 did not predict subsequent outcome of UC but was associated with a significant reduction in the likelihood for colectomy at 1 year [79]. Extended treatment with IFX for up to three additional years was effective in maintaining remission and well tolerated without any additional safety signals [80].

Numerous uncontrolled case series have been reported on patients with UC of varied degrees of severity and refractoriness to prior therapies who were treated with IFX alone or combined with AZA for variable periods of time. Although these data are difficult to subject to group analysis, a nonsystematic review [81] and a systematic review [82] of the literature have confirmed the efficacy and safety of IFX to induce and maintain clinical response, remission, mucosal healing and wane steroids in a considerable proportion of patients with active moderate-to-severe UC.

Colectomy-free survival after IFX therapy was investigated in four studies. Gustavsson et al. [83] reported on a 3-year follow-up of patients who received a single infusion of 4–5 mg/kg IFX as salvage therapy for severe intravenous steroid-refractory UC. Overall, 12/24 (50 %) of IFX-treated patients versus 16/21 (76 %) of placebo-treated patients underwent colectomy (p = 0.012). Lack of mucosal healing at 3 months after the single infusion of IFX predicted subsequent colectomy. In a cohort of 121 patients from Leuven [84] and in a French cohort of 119 patients [85], who were treated with IFX for a median of 33 (IQR 17.0–49.8) months and 18 (IQR 8–32) months, respectively, predictors for colectomy were the lack of short-term response to IFX, baseline levels of CRP ≥5 mg/l [84] or ≥10 mg/l [85], prior treatment with intravenous steroids [84] and/or ciclosporin [84, 85] and an indication of IFX for severe UC [85]. In a Canadian cohort, 46/115 (40 %) of UC patients treated with IFX and/or immunomodulators came to colectomy after a median of 5.3 months. Patients with a detectable serum IFX trough level but not antibodies to IFX had higher rates of remission (69 % vs. 15 %; p < 0.001) and endoscopic improvement (76 % vs. 28 %, p < 0.001). In contrast, an undetectable serum IFX was highly predictive of increased risk for colectomy (55 % vs. 7 %, OR 9.3; 95 % CI 2.9–29.9; p < 0.001). Concomitant use of immunomodulators did not influence the clinical outcomes [86].

In a recent trial involving patients with steroid-refractory UC who were naïve to immunomodulators and anti-TNF therapy, patients were assigned randomly to receive intravenous infusions of infliximab (5 mg/kg at weeks 0, 2, 6 and 14) plus daily oral placebo capsules, oral azathioprine 2.5 mg/kg daily plus placebo infusions on the infliximab schedule or combination therapy with the two drugs. Corticosteroid-free clinical remission was evaluated at week 16 as a primary end point. Corticosteroid-free remission at week 16 was achieved by 39.7 % (31 of 78) of patients receiving infliximab/azathioprine, compared with 22.1 % (17 of 77) receiving infliximab alone (p = 0.017) and 23.7 % (18 of 76) receiving azathioprine alone (p = 0.032). Mucosal healing at week 16 occurred in 62.8 % (49 of 78) of patients receiving infliximab/azathioprine, compared with 54.6 % (42 of 77) receiving infliximab (p = 0.295) and 36.8 % (28 of 76) receiving azathioprine (p = 0.001). Thus, in antitumour necrosis factor-alpha-naive patients with moderate-to-severe UC treated with infliximab plus azathioprine were more likely to achieve corticosteroid-free remission at 16 weeks than those receiving either monotherapy. Combination therapy treatment was associated with better mucosal healing than azathioprine monotherapy.

Whether UC patients in remission on combined AZA and IFX should discontinue AZA is currently unknown. In Crohn’s disease, long-term combination therapy is associated with a higher IFX trough level [87] and lower rate of loss of response to IFX [88] than IFX monotherapy [88]. Whether the full dose of AZA is required to achieve these goals awaits clarification by future trials. However, the benefits of any long-term combination therapy should be balanced against the increased risk for infections and/or malignancies especially in young male patients [84, 8891]. On the other hand, bridging with IFX to AZA monotherapy in AZA-naïve patients may be easier to achieve in UC than in Crohn’s disease. In any case, the decision to stop IFX and continue on AZA should be discussed only for patients in long-standing sustained steroid-free clinical, serologic (normal CRP), endoscopic (mucosal healing) and/or histologic remission who have a detectable IFX trough level. Again, safety issues, patient preferences, adherence, cost and also the risk of no response or allergic reaction upon re-treatment with IFX should be considered in decision-making.


Adalimumab


Adalimumab (ADA) has been approved for the treatment of UC. Initially, preliminary evidence from small trials and retrospective case series suggests that ADA may be effective and safe in achieving and maintaining long-term clinical response or remission, mucosal healing, steroid sparing, improving quality of life and reducing colectomies in patients who have failed almost all prior therapies [9297]. Concomitant use of AZA was the only independent factor predicting response to ADA in one trial [95]. Recently, a large controlled trial was reported in 494 patients with moderate-to-severe UC who had failed treatment with steroids, immunomodulators and/or IFX (40.3 % of the ITT population). Patients were randomised 1:1 to placebo or ADA (n = 248, 160/80 mg sc at week 0 and 2 and then 40 mg every other week). Dose escalation of ADA was allowed. At weeks 8, 52 and both 8 and 52, significantly more patients on ADA achieved clinical remission and response. Significantly more patients with prior IFX failure achieved clinical remission or response on ADA than placebo at week 52 and sustained clinical response at both weeks 8 and 52. The therapeutic gain in steroid-free remission rate at week 52 was only 7.6 % in favour of ADA but was still significantly statistically superior to placebo (5.7 %, p = 0.035). Adverse events were not significantly different between ADA and placebo [98].


Golimumab


Golimumab is a subcutaneously administered fully human anti-TNF antibody, previously approved for the treatment of rheumatoid arthritis, ankylosing spondylitis and psoriatic arthritis. The treatment of ulcerative colitis recently gained regulatory approval for induction and maintenance of remission in patients who have moderate-to-severe ulcerative colitis based on the data in two registration trials. There has been one randomised placebo-controlled trial [Program of Ulcerative Colitis Research Studies Utilizing an Investigational Treatment-Subcutaneous (PURSUIT-SC)] that assessed induction therapy with subcutaneous golimumab in anti-TNF-α-naive patients with moderate-to-severe ulcerative colitis (Mayo score 6–12 points with an endoscopic subscore >2 points) not responding to conventional therapy with oral mesalamine, oral corticosteroids, and AZA/6-mercaptopurine or unable to taper corticosteroids without recurrence of ulcerative colitis activity. The subsequent PURSUIT-M randomised placebo-controlled trial assessed the efficacy and safety of golimumab in maintaining clinical response in patients who responded to induction treatment with golimumab in the preceding PURSUIT-SC trial. Golimumab was shown to be more efficacious than placebo in inducing clinical response, remission and mucosal healing and improving quality of life.

Based upon the date from these two clinical trials, Golimumab is initially administered subcutaneously at the dose of 200 mg at week 0 followed by 100 mg at week 2 and after that every 4 weeks.


Vedolizumab


Vedolizumab is the most recent agent for treatment of ulcerative colitis (and Crohn’s disease) to gain regulatory approval. The efficacy of treatment with vedolizumab in ulcerative colitis was assessed. 374 patients were randomised to either drug or placebo as part of induction. The response for week 6 was measured by the Mayo score and documented mucosal healing.

During induction, 47.1 % of the patients on vedolizumab versus 25.5 % of the patients on placebo achieved remission (p < 0.001). A second cohort of patients received open-label vedolizumab, and responders from both cohorts were included in the maintenance trial that evaluated clinical remission at week 52. Patients were randomised to receive the drug every 4 or 8 weeks or a placebo. A total of 41.8 % of patients maintained remission when receiving medication every 8 weeks compared to 44.8 % who received the drug every 4 weeks; patients who received placebo had maintenance of remission at a rate of 15.9 %. There was a statistically significant difference in maintenance of remission between patients who received the drug every 8 weeks versus placebo (p < 0.001) and those receiving the drug every 4 weeks versus placebo (p < 0.001). Based on this data, the drug was approved for use in adults with moderate-to-severe ulcerative colitis when one or more standard therapies (corticosteroids, immunomodulators or tumour necrosis factor blocker medications) have not resulted in an adequate response.


Additional Therapies



Methotrexate


Several small retrospective case series mostly in patients intolerant or unresponsive to thiopurines have claimed satisfactory results of methotrexate (MTX) for maintenance of remission in UC [55, 99101]. However, a small subtherapeutic trial did not show maintenance efficacy of oral 12.5 mg MTX once weekly since 64 % of the 14 patients receiving MTX compared to 44 % of 18 placebo-treated patients relapsed within 9 months of follow-up (p = 0.25) [102], and a Cochrane systematic review confirmed these negative results [103]. Based on these data, MTX is not currently recommended for maintenance therapy for UC [3]. At present, there is a multicentre trial (ClinicalTrials.gov Identifier: NCT01393405) evaluating the efficacy of methotrexate 25 mg subcutaneously once weekly as a maintenance therapy for patients with ulcerative colitis in remission.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Mar 29, 2017 | Posted by in GASTROENTEROLOGY | Comments Off on Maintenance of Remission in Ulcerative Colitis

Full access? Get Clinical Tree

Get Clinical Tree app for offline access