New Non-anti-TNF-α Biological Therapies for the Treatment of Inflammatory Bowel Disease



Fig. 35.1
Drugs in pipeline for CD (Amiot and Peyrin-Biroulet [1])



A126025_3_En_35_Fig2_HTML.gif


Fig. 35.2
Drugs in pipeline for UC (Amiot and Peyrin-Biroulet [1])



Table 35.1
Treatments discussed in this chapter





























































































































Target

Name

Development in IBD

Mechanism of action

Cytokines
     

 IL-12/IL-23

Ustekinumab

MEDI 2070 (AMG 139)

BI 655066 (risukizumab)

Phase III (CD, UC)

Phase II (CD)

Inhibits p40 subunit

Blocks binding of IL-23 to its receptor

Inhibits the p19 unit

 IL-6

PF-04236921

Phase II (CD)

IL-6 inhibitor

 IL-13

Tralokinumab

QAX576

Bertilimumab

Phase II (UC)

Phase II (CD)

Phase II (CD, UC)

IL-13 receptor antagonist

Inhibits of IL-13

Blocks the activity of eotaxin-1

 IL-17

Vidofludimus

Phase II

Inhibits IL-17 secretion

 IL-21

ATR107

NNC0114-0006

Phase I

Phase II (CD)

Anti-IL-21 receptor antibody

IL-21 inhibitor

Signaling pathways mediated by cytokines
     

 JAK/STAT

Tofacitinib

GLPG0634 (Filgotinib)

Phase III (UC)

Phase II (CD)

Inhibits JAK1 and JAK3

JAK1 inhibitor

 TGF-b

GED0301 (Mongersen)

Phase III/II (CD/UC)

SMAD7 antisense oligonucleotide

Chemokines
     

 IL-10 antagonist

BMS936557 (eldelumab)

Phase II (CD, UC)

CXCL-10 inhibitor

Antiadhesion molecules

Natalizumab

Approved (CD)

α4 integrin antaogmist

Vedolizumab

Approved (CD, UC)

α4β7 integrin antagonist

Etrolizumab

Phase III (CD, UC)

Blocks β7 subunit of α4β7 and αEβ7 integrins

PF-00547659

Phase II (CD, UC)

MAdCAM-1 protein inhibitor

AJM300

Phase III (UC)

α4 integrin antagonist

Alicaforsen

Phase III (UC)

Targets intercellular adhesion molecule 1 (ICAM-1)

AMG181 (abrilumab)

D/c’d

α4β7 integrin antagonist

Firategrast
 
α4 integrin antagonist

GLPG0974

Phase II (UC)

Against FFA2

TRK-170

Phase II (CD)

α4β1/α4β7 integrin antagonist

Anti-inflammatory cytokine

IL-10
 
IL-10 replacement

T-cell stimulation and induction of apoptosis blockades

Laquinimod

DIMS0150

Phase II (CD)

Phase III (UC)

Modulation of immune cells

Activates TLR9

Spingosine-1-phosphate receptor modulators

Fingolimod

APD334

RPC1063 (ozanimod)

Phase II (UC)

Phase III/II (UC/CD)

Traps lymphocytes in lymph nodes




Cytokine Targets



IL-12/IL-23


Interleukin (IL)-12 and IL-23 have been shown to have a central role in the inflammatory pathway in Crohn’s disease, psoriasis and multiple sclerosis [2]. The IL-12 family of cytokines (which includes IL-23) is involved in stimulating innate immunity and developing adaptive immunity. Interleukin-12 and interleukin-23 are key proinflammatory cytokines involved in type 1 helper T (Th1) cell response, which is characterized by a marked accumulation of macrophages making interleukin-12, the major Th1-inducing factor, in Crohn’s disease mucosa [3, 4]. Risk for developing CD and UC has been demonstrated through genome-wide association studies studying variants of the gene encoding the IL-23 receptor and the locus for the gene encoding the p40 chain [5]. It was also suggested that the production of both IL-12 and IL-23 is downregulated in patients with Crohn’s disease but not with ulcerative colitis following administration of IL-12p40 monoclonal antibody [6].

IL-12 is a heterodimer of p40 and p35 subunits. IL-12 induces the differentiation of naïve CD4+ T cells into T helper 1 cells that produce interferon (IFN) gamma and mediates cellular immunity [7]. IL-23 is a heterodimer of the same p40 subunit and a p19 subunit which induces naïve CD4+ T cells into T helper 17 cells which then induce the production of proinflammatory cytokines such as IL-17, IL-6, and TNF-α [8].


Ustekinumab


Ustekinumab (originally known as CNTO 1275) is a human monoclonal antibody (IgG1) that targets the IL-12/23 shared p40 subunit. The result is the inhibition of IL-12 and IL-23 binding to their receptor on the surface of T cells, natural killer cells, and antigen-presenting cells (see Fig. 35.3).

A126025_3_En_35_Fig3_HTML.gif


Fig. 35.3
Ustekinumab mechanism of action (Onuora [9])

Ustekinumab has been shown to be clinically effective in the treatment of moderate to severe CD in phase II studies. There were two groups studied with population 1 being double-blind, placebo-controlled, parallel-group, crossover at week 8 study and population 2 being an open-label study [10]. Both groups were followed up for 28 weeks. Group 1 comprised of 104 patients with moderate to severe Crohn’s disease despite treatment with 5-ASA, antibiotics, corticosteroids, infliximab, and/or immunomodulators who were randomly assigned to one of four arms of treatment: subcutaneous placebo at weeks 0–3 followed by subcutaneous ustekinumab 90 mg at weeks 8–11; subcutaneous ustekinumab 90 mg at weeks 0–3 followed by subcutaneous placebo at weeks 8–11; intravenous placebo at week 0 followed by intravenous ustekinumab 4.5 mg/kg at week 8; or intravenous ustekinumab 4.5 mg/kg at week 0 followed by intravenous placebo at week 8. Group 2 consisted of 27 patients who previously failed to respond to a three-dose induction of infliximab 5 mg/kg or lost response to infliximab maintenance every 8 weeks despite dose escalation to 10 mg/kg who were randomly assigned to open-label therapy with either 4 weekly subcutaneous injections ustekinumab 90 mg at weeks 0–3 or single intravenous ustekinumab 4.5 mg/kg at week 0. The primary endpoint was clinical response at week 8 (a reduction of at ≥25 % and 70 points in the CDAI score from baseline). In group 1 the primary endpoint was assessed for combined subcutaneous and intravenous ustekinumab administered before crossover to placebo (n = 51) versus combined subcutaneous and intravenous placebo administered before crossover to ustekinumab (n = 53). There was no significant difference in rates of clinical response between ustekinumab and placebo at week 8 (49 % vs. 40 %, P = 0.34). However, superiority of ustekinumab over placebo was observed in achieving clinical response at week 4 and at week 6 (53 % vs. 30 % at both time points, P = 0.02). Of note, among patients in group 1 who had prior exposure to infliximab given at submaximal infliximab doses or regimens ustekinumab was significantly superior to placebo with clinical response rates of 59 % and 26 %, respectively (P = 0.022). Among group 2, clinical response rates at week 8 were 43 % and 54 % for subcutaneous and intravenous route of administration of ustekinumab, respectively. It was thus concluded that ustekinumab has the potential of inducing clinical response in patients with moderate to severe active Crohn’s disease with the most prominent effect at weeks 4 through 6 and in patients with prior exposure to infliximab.

Ustekinumab in CD was further studied in a phase IIb randomized, double-blind, placebo-controlled trial with an 8-week induction and 26-week maintenance evaluation period for patients previously exposed to anti-TNF therapy who either had primary nonresponse, secondary nonresponse or intolerance to anti-TNF treatment [11]. During the induction phase, 526 patients were randomly assigned to receive a weight-based dose of intravenous (IV) ustekinumab (1, 3, or 6 mg/kg) or placebo. Responders and nonresponders were separately randomized to subcutaneous (SC) ustekinumab 90 mg or placebo at weeks 8 and 16. The primary endpoint was clinical response (≥100-point decrease from baseline CDAI) at week 6. 39.7 % of patients at 6 mg/kg vs. 23.5 % of patients receiving placebo achieved clinical response at week 6 (P = 0.005). Clinical remission rates at week 6, however, were similar between the ustekinumab and placebo groups. Among patients with a response to ustekinumab in the induction phase, 69.4 % vs. 42.5 % and 41.7 % vs. 27.4 % of patients receiving 90 mg of ustekinumab vs. placebo in the maintenance phase continued to have a clinical response and clinical remission at week 22. In addition, 30.6 % vs. 17.8 % of patients in the ustekinumab vs. placebo group were in glucocorticoid-free remission at week 22 (p = 0.048). Of note, for those patients who did not achieve clinical response after induction with ustekinumab, further therapy with the drug was no different than placebo in terms of clinical response at week 22.

Mucosal healing was also evaluated in the phase IIb trial. Fifty patients were evaluated and 1 out of 9 vs. 8 out of 41 patients achieved mucosal healing in the placebo vs. ustekinumab group respectively (P = 1.00) [11].

Having had promising results with the phase II trials, two phase III trials are completed and a third is underway. UNITI-1 (NCT01369329) is a multicenter, double-blind, placebo-controlled study of ustekinumab for induction of remission in moderate to severe CD in patients with prior failure or intolerance to TNF antagonist therapy [12]. Patients with moderate-severely active CD (defined as a CDAI 220–450) who previously failed or were intolerant to at least 1 TNF-antagonist were randomized (1:1:1) to receive a single dose of IV placebo (PBO), ustekinumab 130 mg, or weight-based tiered ustekinumab dosing approximating 6 mg/kg (260 mg [weight ≤55 kg], 390 mg [weight >55 kg and ≤85 kg], and 520 mg [weight >85 kg]) at week 0. The primary endpoint was clinical response at week 6, defined as reduction from baseline in the CDAI score of >100 points; patients with baseline CDAI score 220–248 were considered to have had a clinical response if a CDAI score of <150 was present. At week 8, patients either transitioned to the UNITI maintenance study (see below) or were followed to week 20. Seven hundred and forty-one patients had had a history of anti-TNF therapy failure either primary nonresponse, secondary nonresponse or intolerance to at least one anti-TNF agent. Primary and secondary endpoints were met at both intravenous doses of ustekinumab. 33.7 % of the ∼6 mg/kg and 34.3 % of the 130 mg ustekinumab groups versus 21.5 % in placebo (P = 0.003 and 0.002, respectively) achieved clinical response at week 6. 20.9 % of the ∼6 mg/kg group and 15.9 % of the 130 mg ustekinumab group versus 7.3 % on placebo (P < 0.001, P = 0.003, respectively) were found to be in clinical remission (CDAI <150) at week 8. 37.8 % of the ∼6 mg/kg and 33.5 % of the 130 mg ustekinumab groups, versus 20.2 % on placebo (each P ≤ 0.001) had a clinical response at week 8. Both intravenous ustekinumab induction doses compared to placebo resulted in significant improvements in CDAI, IBDQ, CRP, fecal lactoferrin, and calprotectin. AEs, SAEs, and infections were similar in the treated and placebo groups. One opportunistic infection (Listeria meningitis) was reported in the ∼6 mg/kg UST group; otherwise no tuberculosis, malignancies, deaths, or major adverse cardiovascular events occurred in ustekinumab treated patients through the week 20 observation.

UNITI-2 (NCT01369342) is identical in design to UNITI-1 and involves patients who have failed conventional treatment (corticosteroids, immunomodulators) or who are corticosteroid dependent without previous failure or intolerance to TNF antagonist therapy. The data were recently presented at the American College of Gastroenterology (Oct 2015 [13]). Six hundred and twenty-eight patients with moderate to severely active Crohn’s disease were randomized to receive a single dose of IV placebo (n = 210), 130 mg of ustekinumab (n = 209) or weight-based tiered dosing of 6 mg/kg of ustekinumab (n = 209). The primary endpoint was clinical response at 6 weeks and at 8 weeks, patients either transitioned to the maintenance study or were followed through to 20 weeks. 51.7 % of patients who received 130 mg of ustekinumab and 55.5 % of patients who received 6 mg/kg of ustekinumab vs. 28 % of patients who received placebo (P < 0.001) achieved a clinical response at 6 weeks. 47 % of patients who received 130 mg of ustekinumab and 58 % of patients who received 6 mg/kg of ustekinumab vs. 32 % of patients who received placebo (P < 0.001) achieved clinical response at week 8. 31 % who received 130 mg of ustekinumab and 40 % of patients who received 6 mg/kg of ustekinumab achieved clinical remission at 8 weeks. Adverse events and serious adverse events were similar between the groups and no malignancies, deaths, opportunistic infections or tuberculosis occurred in patients treated with ustekinumab.

Patients with a clinical response to IV ustekinumab in UNITI-1 or UNITI-2 as mentioned above were eligible for enrollment in the maintenance trial, IM-UNITI (NCT01369355). This study is still ongoing [14]. Patients will be randomized to 90 mg of ustekinumab every 8 weeks, every 12 weeks, or placebo. Patients who receive placebo or ustekinumab every 12 weeks and who lose response are eligible to cross over to receive ustekinumab 90 mg every 8 weeks. The primary outcome will be clinical remission at week 44. Secondary endpoints clinical remission in patients who have previously failed TNF antagonists, clinical response, and corticosteroid-free remission at week 44. Patients who are continuing to do well at week 44 will be eligible to continue to receive ustekinumab in the long-term extension study with follow-up to 3 additional years.


Pediatric Data

Data regarding efficacy of ustekinumab in pediatric CD are lacking. However, Rinawi and colleagues attempted to induce remission with ustekinumab in a 6-year-old patient with active colonic disease, chronic arthritis, and failure to respond to all approved therapies for pediatric CD. The child received three subcutaneous doses of ustekinumab (1.3 mg/kg) at months 0, 1, and 3 which resulted in complete clinical and biochemical remission with no observed adverse events. Clinical response including resolution of diarrhea and arthritis was achieved within the first 2 months of treatment. Furthermore, 1 year after the induction, a weight gain of 3 kg and growth of 8 cm were noted and inflammatory blood markers (CRP, ESR) had normalized and albumin and hemoglobin increased. Remission was maintained with azathioprine 1 year following induction [15].

Bishop and colleagues recently described the use of ustekinumab in four adolescent patients with pediatric Crohn’s disease – ages of initiation were 12, 13, 16, and 17. All patients had previously received corticosteroids, methotrexate, azathioprine/6-mercaptopurine, and both infliximab and adalimumab. Two patients showed clinical response and remained on ustekinumab, but two patients discontinued therapy due to continued symptoms and disease complications and required multiple hospitalizations [16].


Safety

In the phase II trials in Crohn’s disease, there were no increase in the number of adverse or serious adverse events in patients given ustekinumab through week 8 compared with placebo [10]. There were no serious or opportunistic infections. 14 % vs. 8 % of patients in the placebo group vs. drug group reported worsening CD. Three patients (6 %) in the placebo group experienced one or more serious adverse events: small intestinal stenosis and nonsteroidal anti-inflammatory drug–induced gastrointestinal ulceration, worsening CD and erythema nodosum, worsening CD disease and small intestinal obstruction. Two patients (4 %) in the ustekinumab group experienced one or more serious adverse events: small intestinal obstruction and coronary artery disease.

In the patients in the open-label trial followed through week 28, six patients (6 %) in the population 1 group experienced one or more serious adverse events: worsening CD (n = 2), colonic stenosis and pneumothorax (n = 1), small intestinal obstruction (n = 2), and prostate cancer (n = 1). Four patients (15 %) in the population 2 group experienced one or more serious adverse events: viral gastroenteritis (n = 1), nephrolithiasis (n = 1), worsening CD (n = 1), worsening CD, syncope, and disseminated histoplasmosis (n = 1, in a patient receiving prednisone (80 mg/day), azathioprine during the trial and with approximately 3 years prior infliximab use).

In the phase IIb trial, the overall rates of infection were also similar between the ustekinumab and placebo groups during the induction phase [11]. Serious infections were reported in five patients receiving ustekinumab 6 mg/kg (Clostridium difficile infection, viral gastroenteritis, urinary tract infection, anal abscess, and vaginal abscess). In one patient receiving ustekinumab 1 mg/kg, there was central catheter infection with Staphylococcus, and in one patient receiving placebo, there was an anal abscess. Infusion reactions were similar between the ustekinumab and the placebo groups.

In the maintenance phase, there were no deaths, major adverse cardiovascular events, tuberculosis, or other serious opportunistic infections. One patient who received 1 mg/kg ustekinumab followed by 90 mg of ustekinumab was reported to have basal-cell carcinoma. The most commonly reported adverse events in the maintenance phase, other than those related to CD, were nonserious infections (39.2 %), abdominal pain (7.2 %), nausea (6.1 %), and nasopharyngitis (6.1 %) in the ustekinumab group and these were all numerically lower than events in the placebo group, except for the rate of nasopharyngitis (3.3 %).

The long-term safety profile of ustekinumab has been evaluated in the treatment of psoriasis. Pooled safety data from four clinical trials of ustekinumab for psoriasis revealed a total of 3117 patients received at least one dose of ustekinumab, with a total of 8998 person-years of follow-up, with at least 4 years of exposure in 1482 patients [17]. At year 5, events per 100 patient years of follow-up for ustekinumab 45 mg and 90 mg were as follows: serious adverse events was 7.0 and 7.2, serious infections 0.98 and 1.19, nonmelanoma skin cancers 0.64 and 0.44, and major adverse cardiovascular events (MACE) including myocardial infarction, cerebrovascular accident or cardiovascular death 0.56 and 0.36. Adverse events or rates of overall mortality and other malignancy did not seem to increase over time compared with an age-matched and sex-matched US population.

There has been one case report of central demyelination in 63-year-old woman with CD. This patient had previous exposure to immunomodulator therapy, infliximab, adalimumab, and certolizumab and received ustekinumab through the clinical trial program and afterwards for compassionate reasons. About 1 year after first receiving ustekinumab, she developed progressive weakness and was diagnosed with primary progressive multiple sclerosis [18].


AMG139 (MEDI2070)


In contrast to IL-12 inhibition, IL-23 inhibition has been previously shown to be associated with a decreased incidence of tumor formation [19]. In addition, there have been concerns regarding the safety of ustekinumab and/or briakinumab regarding the incidence of serious infections and major adverse cardiovascular events [19]. Therefore, it is thought that IL-23-specific antagonism may provide similar or greater efficacy than blocking IL-12/23p40 and without the potential risks associated with blocking IL-12. Accordingly, a human IgG2 monoclonal antibody called AMG139 was developed to specifically bind to the IL-23 p19 unit and block binding of IL-23 specifically to its receptor. A phase I trial was just completed assessing the safety and tolerability of AMG139 following the administration of multiple intravenous (IV) or subcutaneous (SC) doses in healthy subjects and in subjects with mild to severe CD [20].

Phase IIb studies are ongoing at the writing of this chapter.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of AMG139 in children or adolescents with IBD.


BI 655066


BI 655066 is a monoclonal antibody that also targets IL-23p19. It is being investigated as a subcutaneous drug in both psoriasis and Crohn’s disease. A proof of concept, multicenter, randomized, double-blind, placebo-controlled, parallel-group phase II dose-ranging study of BI 655066 in patients with moderately to severely active Crohn’s disease is ongoing. The primary outcome measure is clinical remission at week 12, defined as a CDAI score of <150. Secondary outcome measures include CDEIS remission, CDEIS response, mucosal healing, deep remission, and clinical response at week 12 [21]. There is another ongoing an open-label, single-group, long-term safety extension trial ongoing for subjects who responded to treatment with BI 655066 in a preceding trial [22].


Pediatric Data

At the time of writing this chapter, there are no published data on the use of BI 655066 in children or adolescents with IBD.


Safety

Selectively blocking IL-23 vs. IL-12 has been thought to be more targeted with less side effects as a result [23].

A single-rising dose, multicenter, randomized, double-blind, placebo-controlled, within-dose phase I trial was the first-in-human proof-of-concept study and in this study the clinical and biological effects of BI 655066 was evaluated in patients with moderate to severe plaque psoriasis. Patients received 0.01, 0.05, 0.25, 1, 3, or 5 mg/kg BI 655066 intravenously, 0.25 or 1 mg/kg BI 655066 subcutaneously, or matched placebo. Thirty-nine patients received single-dose BI 655066 intravenously (n = 18) or subcutaneously (n = 13) or placebo (n = 8). BI 655066 was well tolerated. The number of adverse events were similar between the BI 655066 and placebo groups. Four serious adverse events were reported among BI 655066-treated patients and were thought not to be treatment related [24].


IL-6


Interleukin-6 (IL-6) is a cytokine with central roles in immune regulation, inflammation, hematopoiesis, and oncogenesis. It is a contributor of Th-17 differentiation [1]. Increased levels of IL-6 and soluble IL-6 receptor have been demonstrated in both serum and intestinal tissues of the patients with active Crohn’s disease and with a more severe form [25].


PF-04236921


PF-04236921 is a monoclonal antibody against IL-6. A phase II placebo-controlled study has been completed to evaluate the safety and efficacy of this subcutaneously administrated antibody in patients with active CD (the ANDANTE study) [26]. There were limitations of the study including early termination which led to small numbers of participants and technical problems with measurement resulting in unreliable or uninterpretable data.

Phase I/II studies are ongoing at the time of writing of the chapter.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of PF-04236921 in children or adolescents with IBD.


IL-13


Interleukin-13 (IL-13) is a central cytokine in the T helper 2 immune response [2729]. IL-13 has effects on many cell types including B cells, monocytes, macrophages, epithelial cells, smooth muscle cells and neurons and has been indicated in the pathogenesis of many disease including asthma, scleroderma in addition to IBD [30]. Its upregulation has been proposed to be a key driver of mucosal inflammation – specifically in UC.


Tralokinumab


Tralokinumab (CAT-354) is an IL-13-specific human immunoglobulin G4 monoclonal antibody that binds to and neutralizes IL-13 [31, 32]. Tralokinumab is being evaluated in phase III clinical studies in patients with asthma and in a phase II study in idiopathic pulmonary fibrosis and may be a treatment for UC.

In a phase IIa, randomized, double-blind, placebo-controlled, parallel-group, multicenter trial, 111 patients with UC (total Mayo score ≥6) were randomized to tralokinumab 300 mg subcutaneous or placebo [33]. The primary endpoint of clinical response at week 8 was 38 % (21/56) for tralokinumab vs. 33 % (18/55) for placebo (P = 0.406). Clinical remission rate at week 8 was 18 % (10/56) vs. 6 % (3/55) (P = 0.033) and mucosal healing rate was 32 % (18/56) vs. 20 % (11/55) (P = 0.104) for tralokinumab vs. placebo.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of tralokinumab in children or adolescents with IBD.


Safety

Tralokinumab had an acceptable safety profile in the only phase IIa study to date [33]. The median duration of exposure was 84 days. The number of patients who experienced adverse events was similar in the tralokinumab and placebo groups. The most frequently reported adverse events were symptoms of UC and headache. The number of patients discontinuing treatment because of adverse events was similar in both groups and the most common adverse event leading to discontinuation was symptoms of UC.


QAX576


QAX576 is a highly potent and specific inhibitor of human IL-13 activity in cell-based in vitro assays. It has been used to treat eosinophilic esophagitis with success [34] and studied in asthma and idiopathic pulmonary fibrosis.

A phase II study to assess the safety and efficacy of intravenously administered QAX576 in patients with fistulizing Crohn’s disease has been completed [35]. Another phase II study to test the safety and efficacy of the drug in the treatment of perianal fistulas has also been completed [36]. Results are not available of either of the studies.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of QAX576 in children or adolescents with IBD.


Bertilimumab


Bertilimumab is a fully human, IgG4-type monoclonal antibody that blocks the activity of a protein called eotaxin-1. Eotaxin-1 plays an important role in inflammation and causes eosinophils to migrate towards sites of inflammation where they become activated and release substances that result in tissue damage and enhance inflammation.

A randomized, double blind, placebo-controlled, parallel-group, multicenter study in adult patients with active moderate to severe UC is ongoing. Patients are currently being and eligible patients will be randomly assigned in a 2:1 ratio to one of two treatment groups, bertilimumab 10 mg/kg intravenously or matching placebo, respectively [37].


Pediatric Data

At the time of writing this chapter, there are no published data on the use of Bertilimumab in children or adolescents with IBD.


IL-17



Vidofludimus


Vidofludimus (4SC-101) is a novel oral immunomodulatory drug that inhibits dihydro-orotate dehydrogenase and lymphocyte proliferation in vitro and inhibits interleukin (IL)-17 secretion in vitro independently of effects on lymphocyte proliferation [38].

A phase IIa open-label, single-arm trial with vidofludimus (ENTRANCE trial) in inflammatory bowel disease was performed. The primary outcome was to assess remission-maintenance potential in steroid dependent IBD patients upon steroid weaning (ECCO 2011). There were 26 CD and UC patients. There was an 88.5 % response rate of both complete and partial responders in patients on vidofludimus vs. 20 % placebo. 53.9 % (14/26) patients were complete responders, 34.6 % (9/26) patients were partial responders and 11.5 % (3/26) patients were nonresponders. There was no difference in response rates between Crohn’s disease (85.7 %) and ulcerative colitis (91.7 %). In addition, the average prednisolone consumption dramatically dropped during treatment with the drug. All 26 patients reached a relapse-free prednisolone dose which was significantly (p < 0.001) lower than their individual threshold doses at which they experienced relapses prior to entering the study. Mean prednisolone consumption was significantly lowered from 26.5 mg/day (±8.0) at the start of treatment to 1.0 mg/day (±2.7) at week 12.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of vidofludimus in children or adolescents with IBD.


Safety

Vidofludimus was safe and well tolerated by all patients in the ENTRANCE trial. A total of 75 adverse events were reported (53 mild, 18 moderate, 4 severe) of which 19 adverse events were judged as possibly or probably drug related and included nasopharyngitis, abdominal pain, fatigue, insomnia, glucosuria, leucocyturia, microhematuria, musculoskeletal pain, myalgia, tachycardia, and dyspepsia. No drug related serious adverse events were reported.


IL-21



ATR-107


ATR-107 is a fully human anti-IL-21 receptor (IL-21R) monoclonal antibody designed to block IL-21 from binding and activating the receptor as a novel approach to treatment of systemic lupus erythematosus and other autoimmune diseases [3941]. The ATR-107 target, IL-21R, is expressed on many lymphoid cells, including B cells, activated T cells, natural killer cells, monocytes and dendritic cells and thus ATR-107 is expected to block the effects of IL-21 activation of its receptor (enhanced proliferation of lymphoid cells, B cell differentiation to memory cells and plasma cells, and development of T helper type 17 (Th17) cells) [42, 43]. In addition, anti-inflammatory efficacy resulting from IL-21R blockade has been observed in animal models [44].

Phase I studies are ongoing at the writing of this chapter.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of ATR-107 in children or adolescents with IBD.


NNC0114-0006


NNC0114-0006 is an anti-IL-21-antibody. A randomized, double-blind, placebo-controlled, parallel-group trial phase II study to assess clinical efficacy and safety of NNC0114-0006 in subjects with active Crohn’s disease has been completed. Results are not yet known.


Pediatric Data

At the time of writing this chapter, there are no published data on the use of NNC0114-0006 in children or adolescents with IBD.


Blockade of the Downstream Signaling Pathways Mediated by Cytokine



JAK/STAT Pathway


Janus kinases (JAK) 1, 2 and 3 are extremely important in cytokine signaling that is involved in lymphocyte survival, proliferation, differentiation and apoptosis [45]. JAK3 is found only in hematopoietic cells and is part of the signaling pathway activated by IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 which is crucial in the activation, function and proliferation of lymphocytes [46] (see Fig. 35.4).

A126025_3_En_35_Fig4_HTML.jpg


Fig. 35.4
JAK pathway inhibitors (Neurath [47])


Tofacitinib


Tofacitinib (CP-690,550) is an oral small molecule inhibitor of JAK 1 and 3. In vitro studies have shown that it interferes with Th2 and Th17 cell differentiation and blocks the production of IL-17 and IL-22 [48].

In a phase II, double-blind, placebo-controlled trial, the efficacy of tofacitinib in 194 adults with moderately to severely active ulcerative colitis was evaluated [49]. Patients were randomly assigned to receive tofacitinib at a dose of 0.5 mg, 3 mg, 10 mg, or 15 mg or placebo twice daily for 8 weeks. The primary outcome was a clinical response at 8 weeks and occurred in 32 %, 48 %, 61 %, and 78 % of patients receiving tofacitinib at a dose of 0.5 mg (P = 0.39), 3 mg (P = 0.55), 10 mg (P = 0.10), and 15 mg (P < 0.001), respectively – compared with 42 % of patients receiving placebo. Clinical remission (Mayo score ≤2 with no subscore >1) at 8 weeks occurred in 13 %, 33 %, 48 %, and 41 % of patients receiving tofacitinib at a dose of 0.5 mg (P = 0.76), 3 mg (P = 0.01), 10 mg (P < 0.001), and 15 mg (P < 0.001), respectively, as compared with 10 % of patients receiving placebo. Treatment with the drug resulted in reduced C-reactive protein and fecal calprotectin levels.

Tofacitinib was also evaluated in patients with moderate to severe active CD. Patients were randomized to receive tofacitinib twice daily for 4 weeks at doses of 1 mg, 5 mg, 15 mg, or placebo [50]. The primary endpoint was not met in this phase II trial in CD patients receiving tofacitinib, but the placebo response rate was high. Although no significant clinical differences were observed between actively treated patients and placebo, a reduction in CRP and fecal calprotectin levels among patients given 15 mg tofacitinib twice daily indicated its biologic activity. The primary endpoint was clinical response at week 4 and the rates were as follows: 36 % (P = 0.467), 58 % (P = 0.466), and 46 % (P ≥ 0.999) in those patients given 1, 5, or 15 mg tofacitinib twice daily versus 47 % given placebo. A secondary endpoint was clinical remission at week 4 and this was seen in 31 % (P = 0.417), 24 % (P = 0.776), and 14 % (P = 0.540) of patients given 1, 5, and 15 mg tofacitinib, versus 21 % of patients given placebo. As the clinical response was not significant, the trial was negative. However, the placebo response and remission rates were unexpectedly high and in addition, the reduction in fecal calprotectin and C-reactive protein levels among patients receiving 15 mg tofacitinib twice daily suggested biologic activity of the drug. A repeat phase II trial with stricter inclusion criteria defining active Crohn’s disease is ongoing [51].

There is currently an ongoing multicenter, randomized, double-blind, placebo-controlled, parallel-group study evaluating tofacitinib as a maintenance therapy in patients with ulcerative colitis. Patients will either be given placebo orally twice daily, tofacitinib 5 mg orally twice daily or tofacitinib 10 mg orally twice daily. The primary endpoint is the proportion of subjects in remission at week 52. Secondary outcomes that will be measured is the proportion of patients with mucosal healing at week 52 and number of patients in sustained steroid free remission [52].


Pediatric Data

At the time of writing this chapter, there are no published data on the use of tofacitinib in children or adolescents with IBD.


Safety

Tofacitinib has generally been well tolerated in clinical trials. The most commonly reported adverse events related to infection reported by Sandborn and colleagues were influenza and nasopharyngitis [49, 50]. Two patients receiving the 10 mg dose twice daily had serious adverse events from infection (postoperative abscess, anal abscess). Of significance but uncertain long term consequence, a dose-dependent increase in low-density and high-density lipoprotein cholesterol was seen after 8 weeks of treatment which were reversible after discontinuing the study drugs [49, 50]. It is not clear what the long-term effects of this is. Three patients treated with tofacitinib (one at dose of 10 mg twice daily and two at dose of 15 mg twice daily) had an absolute neutrophil count of less than 1500 (with none being <1000) [53, 54]. Tofacitinib is a true immunosuppressant and there is a concern for increased risk of infections and lymphoma with using this drug compared to other biologics. However, this drug can be used as monotherapy which makes it a very appealing option. Herpes zoster infections have been observed quite frequently and in rheumatoid arthritis, several cases of lymphoma were reported, but the overall risks of infections and mortality with tofacitinib seem to be similar to those observed with other biologic agents [51].

This drug is administered orally and can be used as monotherapy, which makes it a very appealing option.

Other JAK inhibitors are currently under clinical investigation in phase II for both CD and UC [51].


GLPG0634


GLPG0634, has been shown to selectively inhibit JAK1-dependent signaling in cellular and whole blood assays as well as showed remarkable efficacy in collagen induced arthritis disease models for RA in both mouse and rat [55].

There is currently an ongoing phase II clinical trial evaluating the efficacy, safety and tolerability and pharmacokinetics of GLP0634 on Crohn’s disease during a 20-week period of time [56].


Pediatric Data

At the time of writing this chapter, there are no published data on the use of GLPG0634 in children or adolescents with IBD.


TGF-B


One mechanism by which Crohn’s disease develops involves transforming growth factor (TGF)-b which is a suppressive cytokine [57, 58]. SMAD7 is an endogenous inhibitor of the immunosuppressive cytokine transforming growth factor-β1. In Crohn’s disease, TGF-b1 activity is inhibited by high Smad7, an intracellular protein that binds to the TGF-b1 receptor and prevents TGF-b1-driven signaling [59, 60]. Studies in mice have consistently shown that the induction of experimental CD-like colitis is associated with enhanced expression of Smad7 and reduced TGF-b1 activity [59]. The inhibition of Smad7 in CD mucosal cells with a specific antisense oligonucleotide has been demonstrated to restore TGF-b1 activity which therefore down-regulates the production of inflammatory cytokines [61].


GED0301 (Mongersen)


GED0301 is an antisense oligonucleotide targeting SMAD7 and is an oral gastro-resistant compound with a pH-dependent, delayed-release of the oligonucleotide in the terminal ileum and right colon.

A phase I clinical trial was performed which showed that GED0301 in active, steroid dependent/resistant CD patients resulted in a clinical benefit in all patients [62, 63].

In a placebo-controlled phase II study in patients with active CD [64], patients were randomized to receive induction treatment with different doses of GED0301 (Mongersen) or placebo for 2 weeks. The primary endpoint was clinical remission and this was seen in 55.0, 65.1, and 9.5 % of patients receiving Mongersen 40 mg/day, 160 mg/day, or placebo (p < 0.0001, for both comparisons) at 15 days and maintained for ≥2 weeks. Adverse events were similar across the treatment groups.

This promising molecule is now being investigated in larger trials [51].


Pediatric Data

At the time of writing this chapter, there are no published data on the use of GED0301 in children or adolescents with IBD.


Safety

A phase I clinical trial using GED0301 in active, steroid dependent/resistant CD patients was safe and well tolerated [62]. Adverse events were similar across the treatment groups in a phase II clinical trial [64].


Targeting Chemokines


Chemokines are cytokine proteins expressed in lymphoid and nonlymphoid tissue, thought to be involved in leukocyte trafficking. Their effects are mediated by g-protein coupled transmembrane receptors, which are classified by cysteine residues. Persistent, aberrant leukocyte chemotaxis to inflamed mucosa is thought to play a role in the pathogenesis of inflammatory bowel disease. Increased expression of several chemokines has been reported in patients with ulcerative colitis and Crohn’s disease.


IP-10 Antagonists


Interferon-γ-inducible protein-10 (IP-10 or CXCL10) is a chemokine that plays an important role in the migration of cells into sites of inflammation by influencing activation and migration of activated T-cells, monocytes, eosinophils, natural killer, epithelial and endothelial cells [65, 66].

The receptor for CXCL-10 is CXCR3 but IL-10 also seems to modulate cellular function independently of CXCR3 [65]. CXCL10 has been found to be expressed in higher levels in the colonic tissue and plasma of patients with UC [67, 68]. In animal models of UC, blocking CXCL10 has been shown to modify disease progression [53, 66, 6971].


BMS936557 (MDX-1100, Eldelumab)


BMS-936557 (Eldelumab) is a fully human, monoclonal antibody to CXCL10. A phase I, open-label, dose-escalation study of MDX-1100 was been performed in patients with UC using MDX-1100 [54]. The primary objective of this was to evaluate the safety of single doses of the drug in patients with ulcerative colitis having exacerbation on stable doses of standard therapy. Patients were off anti-TNF therapy for at least 8 weeks prior to the study. Cohorts of patients were given a single infusion of the drug at doses of 0.3, 1.0, 3.0, or 10 mg/kg and were followed for at least 70 days post infusion. A clinical response was defined as UCDAI decrease by ≥3 points at day 29 compared to baseline. Patients who responded were allowed to receive up to three additional infusions at the time of relapse. Peripheral blood mononuclear cells and colon biopsy specimens were studied for expression of CXCL10 and CXCL10-induced proteins. Three patients in the 1.0 mg/kg and two patients in the 3.0 mg/kg cohorts had clinical responses; however, one patient in the 1.0 mg/kg cohort also was started on concomitant immunomodulator therapy 2 months prior to MDX-1100 administration. Two of three responding patients who relapsed after 50, 85, and 93 days, respectively who had then been given additional MDX-1100 doses responded to re-treatment. One patient in the 3.0 mg/kg cohort was admitted to the hospital for anemia and worsening UC requiring a colectomy but otherwise the drug was well tolerated.

Mayer, and colleagues in 2014 published data from an 8-week phase II, double-blind, multicenter, randomized study in patients with active ulcerative colitis [65]. Patients with moderately to severely active UC were given either BMS-936557 (10 mg/kg) or placebo intravenously every other week. The primary endpoint was the rate of clinical response at day 57 and secondary endpoints were clinical remission and mucosal healing rates. Fifty-five patients received the drug and 54 patients received placebo. Primary and secondary endpoints were not met. However, what was found was that with higher steady-state trough levels of BMS-936557 (108–235 μg/ml), there was an increased clinical response (87.5 % vs. 37 % p < 0.001) and histological improvement (73 % vs. 41 % P = 0.004) compared to placebo. Infections occurred in 12.7 % of BMS-936557 treated patients and 5.8 % placebo-treated patients. Two patients (or 3.6 %) discontinued due to adverse events.

In a phase IIb study of patients with ulcerative colitis [65, 72] patients (n = 121) with CDAI ≥220 and ≤450 were randomly assigned 1:1:1 to placebo or intravenous eldelumab 10 or 20 mg/kg given on days 1 and 8 and then every other week. Endoscopy videos were collected for all patients at baseline. Patients with a score of 2–3 on the ulcerated surface subscore of the Simplified Endoscopic Score for Crohn’s Disease (SES-CD) in at least 1 of 5 segments had a follow up endoscopy at 11 weeks. A central reader read endoscopies in a blinded fashion. Trial endpoints included week 11 clinical response which was defined as a reduction in CDAI 100 points from baseline or an absolute CDAI score <150, clinical remission which was defined as CDAI <150 and endoscopic improvement. There was a trend towards efficacy as remission and response rates at week 11 for the 10 mg/kg dose, 20 mg/kg and placebo groups were 22.5 and 47.5 %, 29.3 and 41.5 %, vs. 20 and 35 % and were higher in anti-TNF-naive patients versus those patients who experienced anti-TNF failures. Both drug groups achieved a greater reduction from baseline in mean endoscopy scores compared to placebo and were similar in the eldelumab-treated groups across the anti-TNF-naive and anti-TNF failure subgroups [73].


Pediatric Data

At the time of writing this chapter, there are no published data on the use of BMS936557 in children or adolescents with IBD.


Safety

Serious adverse events were more common in the eldelumab groups (7.5 and 9.8 %) compared with placebo (5 %) in a phase IIb study of patients with ulcerative colitis [65, 73, 72].


Antiadhesion Molecules



Natalizumab


Natalizumab is a humanized IgG4 monoclonal antibody against the adhesion molecule α4 integrin, which is involved in migration of leukocytes across the endothelium, and is upregulated in sites of inflamed endothelium. It is administered intravenously every 4 weeks. The efficacy of natalizumab for the treatment of multiple sclerosis has been demonstrated in controlled trials [74, 75, 76]. In November 2004 natalizumab was approved by the Food and Drug Administration (FDA) for the treatment of multiple sclerosis with subsequent withdrawal from the market in February 2005, and then reintroduction with certain restrictions for the treatment of multiple sclerosis in September 2006.

Six randomized, double-blind, placebo-controlled trials assessed the efficacy of in patients with Crohn’s disease, whereas only one uncontrolled pilot study has been conducted in patients with ulcerative colitis.

An initial phase IIa trial comprised of 30 patients with active Crohn’s disease who received randomly allocated single infusion of either natalizumab 3 mg/kg or placebo and had CDAI evaluated 2 weeks after infusion [77]. There was a significant decrease in baseline CDAI score in patients treated with natalizumab at week 2 (mean drop 45 points, P = 0.02), while no significant decrease was observed in placebo arm (mean drop 11 points, P = 0.2). It demonstrated a higher rate of clinical remission at week 2 in patients given natalizumab 3 mg/kg compared to placebo (39 % vs. 8 %, P = 0.1) [74]. There was no significant difference between natalizumab and placebo in achieving remission at week 2 (39 % vs. 8 %, P = 0.1). This pilot trial did not show any significant superiority of natalizumab over placebo in treating patients with Crohn’s disease.

The second trial compared natalizumab (single infusion of 3 mg/kg, two infusions of 3 mg/kg or two infusions of 6 mg/kg) versus placebo in 248 patients with moderate to severe Crohn’s disease [78]. The primary endpoint (remission, CDAI < 150 points at week 6) rates were 29 % for single infusion of natalizumab (P = 0.757), 44 % for two infusions of natalizumab 3 mg/kg (P = 0.030) and 31 % for two infusions of natalizumab 6 mg/kg (P = 0.533) versus 27 % in placebo arm. Only two doses of natalizumab 3 mg/kg had statistically significant superiority over placebo in achieving clinical remission.

Three Phase III trials have been conducted in Crohn’s disease. In Efficacy of Natalizumab as Active Crohn Therapy (ENACT-1), 905 patients with moderate to severe Crohn’s disease were randomly assigned to receive induction therapy at weeks 0, 4, and 8 with either natalizumab 300 mg or placebo [79]. The primary endpoint in the induction trial was clinical response defined as at least 70-point decrease in baseline CDAI score at week 10 and it was achieved in 56 % and 49 % of natalizumab and placebo recipients, respectively (P = 0.05) [79]. In ENACT-2, 339 patients who had a response to natalizumab in induction ENACT-1 trial at both week 10 and 12 were randomly reassigned to receive 300 mg of natalizumab or placebo every 4 weeks from week 12 through week 56 [79]. The primary endpoint in ENACT-2 trial was a sustained response through week 36. Patients with at least 70-point increase in CDAI score after week 12 with an absolute CDAI score of at least 220 or needed therapeutic intervention after week 12 were considered losing response. Rates of sustained response at week 36 were 61 % in patients receiving maintenance treatment with natalizumab and 28 % in those receiving placebo maintenance (P < 0.001). It was demonstrated that maintenance treatment with natalizumab is significantly superior to placebo in patients who responded to induction treatment. Concomitant immunosuppressants did not improve the rates of clinical remission or response [80]. Patients who maintained remission on natalizumab over 12 months in the ENACT-2 trial were enrolled into a subsequent phase III, open-label, 2-year open-label extension trial designed to assess long-term efficacy and safety of natalizumab [81]. This open-label trial comprised of 146 patients who received 12 natalizumab infusions over 12 months. The proportion of patients who maintained remission after 6 (week 24) and 12 (week 48) additional infusions of natalizumab was 89 % and 84 %, respectively. This open-label extension trial supported data from ENACT-2 trial that natalizumab maintains remission over additional 12 months in patients with sustained remission on natalizumab in the preceding 12 months.

In the ENCORE trial, 509 patients with moderate to severe Crohn’s disease with elevated C-reactive protein concentrations at baseline were randomized to receive natalizumab 300 mg or placebo at weeks 0, 4, and 8 [82]. Natalizumab was significantly superior over placebo in inducing remission at week 8 that was sustained through week 12 (primary endpoint defined as at least 70-point decrease in CDAI score) with respective proportions of patients of 48 % vs. 32 % (P < 0.001). In addition natalizumab was also significantly superior over placebo in achieving additional efficacy endpoint, namely clinical remission at week 8 sustained through week 12 CDAI <150 that was observed in 26 % of natalizumab-treated patients and 16 % of placebo recipients (P = 0.002).

Finally, Sands et al. performed a placebo-controlled trial in which 79 patients with active Crohn’s disease during ongoing treatment with infliximab 5 mg/kg every 8 weeks for at least 10 weeks before initiation of randomization were randomly assigned to receive three intravenous infusions of either natalizumab 300 mg or placebo every 4 weeks while continuing their initial infliximab regimen during duration of the trial [83]. At week 6 patients treated with natalizumab plus infliximab experienced mean decrease in their CDAI score of 37.7 points, while those treated with placebo plus infliximab experienced small increase in CDAI score of a mean of 3.5 points (P = 0.084). A trend towards greater efficacy of combined treatment with natalizumab and infliximab over infliximab alone was shown in patients with active Crohn’s disease not responding to infliximab therapy.

Gordon et al. published results of one small open-label study of 10 patients with active ulcerative colitis who were treated with a single infusion of natalizumab 3 mg/kg [84]. All patients had their disease activity evaluated using Powell-Tuck score 2 weeks after infusion. Treatment with natalizumab resulted in significant decrease in median disease activity score from 10 at baseline to 6 at 2 weeks postinfusion (P = 0.004). It was suggested that future randomized, placebo-controlled trials are warranted to further assess the efficacy of natalizumab in ulcerative colitis.

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

Stay updated, free articles. Join our Telegram channel

Nov 20, 2017 | Posted by in GASTROENTEROLOGY | Comments Off on New Non-anti-TNF-α Biological Therapies for the Treatment of Inflammatory Bowel Disease

Full access? Get Clinical Tree

Get Clinical Tree app for offline access