• No results found

University of Groningen Complications and optimalisation of Mesalazine and anti-TNF-alpha therapy in inflammatory bowel disease Buurman, Dorien Joke

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Complications and optimalisation of Mesalazine and anti-TNF-alpha therapy in inflammatory bowel disease Buurman, Dorien Joke"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Complications and optimalisation of Mesalazine and anti-TNF-alpha therapy in inflammatory

bowel disease

Buurman, Dorien Joke

DOI:

10.33612/diss.98535663

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Buurman, D. J. (2019). Complications and optimalisation of Mesalazine and anti-TNF-alpha therapy in

inflammatory bowel disease. Rijksuniversiteit Groningen. https://doi.org/10.33612/diss.98535663

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Introduction

(3)

General introduction and outline of this thesis

Inflammatory Bowel Disease (IBD) is a chronic inflammatory disease of the gut that

is characterized by a heterogeneous presentation. It comprises both Crohn’s disease

(CD) and Ulcerative Colitis (UC). IBD is characterized by relapsing symptoms as

diarrhea, abdominal pain, fatigue and weight loss. The diagnosis is confirmed by a

combination of clinical, endoscopical, histological and radiological findings.

1

UC

is a superficial mucosal inflammation that starts in the rectum and may affect the

whole colon. Histological the inflammation is confined to the mucosal layer usually

with a mixed cellular infiltrate without granulomas. As UC only affects the colon CD

can affect the whole gastrointestinal tract. The inflammation is not continuous but

discontinuous. The disease location is most frequent in the colon in combination

with the ileum (41%) but it can also be present in the terminal ileum (37%) or colon

(22%) only.

2

The upper gastrointestinal tract is affected in 10% of the cases.

2

The

ulcers can penetrate the perianal region leading to perianal abscesses or fistula. The

inflammation is transmural, and can contain granuloma’s. Apart from the intestinal

inflammation IBD can also have extra intestinal manifestations in skin (10%), joints

(22%) and eyes (4%).

2

The precise etiology of IBD is unknown, and there is no medical therapy that

can cure IBD. Removal of the colon is a surgical option to cure UC. Current medical

therapy (figure 1) is aiming to control the intestinal inflammation by inducing and

maintaining remission.

3, 4

The treatment strategy for IBD is mainly based on the

severity, distribution and pattern of disease. Figure 1 shows the rounded or range of

reported clinical remission rates upon induction therapy.

3, 4

(4)

Chapter

Part I of this thesis focuses on mesalazine (5-Amino Salicylic Acid, [5-ASA]) which is

the cornerstone of medical treatment of UC. They are highly effective for inducing and

maintaining remission in UC at doses ≥ 2 gram.

5

5-ASA is a relatively safe drug and

has fewer side effects than the original sulfasalazine.

6

Different dosages and enteral

delivery formulations has been developed with no clear difference in efficacy.

7-9

It is

acknowledged, that once-daily dosing is likely to improve compliance ; ≥ 2g/day oral

5-ASA induces remission more effectively than lower doses [relative risk for failure

to achieve remission at weeks 4 – 8 of 0.91, 95% confidence interval 0.85–0.98].

5

Patients with moderate disease may benefit from the higher dose of 4.8 g/day.

3, 8

Although the frequency of adverse events in clinical trials in UC and CD patients were

comparable between 5-ASA and placebo, about 6.5% of patients using 5-ASA develop

adverse effects.

10

The most common adverse effects are nausea, vomiting, headache,

abdominal pain, rash and diarrhea. Adverse effects can be classified into predictable

(type A) and unpredictable (type B) reactions. Type B reactions are responsible for

10-15% of all drug side effects. In some forms of type B reactions, especially in case

of immunologic/allergic reactions, mast cell associated (type-1) desensitization can

be achieved. Desensitization is the procedure to induce tolerance to drugs responsible

for hypersensitivity reactions using a slowly incremental dose of the drug. Effective

desensitization is described for cytostatics, antibiotics and also for sulfasalazine.

11, 12

As

the use of 5-ASA can be limited by the occurrence of adverse events, desensitization

is of interest.

11, 12

A few case-reports reported successful desensitization for patients

intolerant for 5-ASA. In these cases, patients had a variety of side-effects, e.g. urticaria,

fever, exanthema, diarrhea and nausea, sometimes combined with eosinophilia.

13-18

We tried to desensitize UC patients with a demonstrated 5-ASA intolerance with a

rapid desensitization protocol (Chapter 2). Other rare, but severe, adverse side-effects

are hepatotoxicity, pancreatitis, pneumonitis, and interstitial nephritis.

19-22

Patients

with IBD are also at risk for hepatobiliary disease; IBD patients can develop primary

sclerosing cholangitis (PSC) (2%), auto Immune Hepatitis (AIH) and several overlap

syndromes. Therefore, the diagnosis can be challenging. Studies showed that patients

with IBD may also develop acute or chronic hepatic injury as a result of drugs as

5-ASA.

23, 24

We describe a patient with therapy refractory UC who developed a 5-ASA

luxated auto-immune hepatitis/PSC overlap syndrome (Chapter 3).

Nephrotoxicity is a rare idiosyncratic reaction to 5-ASA therapy. Nephrotoxicity

associated with 5-ASA agents was first described in case reports and has since then

been reported multiple times for both sulfasalazine and the more modern 5-ASA

agents.

25, 26

Data from clinical trials suggest an annual risk of 0.26% and data from a

questionnaire sent to gastroenterologists estimated an incidence of 1 case per 4000

patient years.

27, 28

The aim of Chapter 4 was to characterize the clinical features of

this serious adverse event and then perform the first genome wide association study

to identify genetic risk factors for the development of a drug-induced interstitial

nephritis.

In Part II of these thesis we focus on agents against tumor necrosis factor alpha

(TNF-alpha) in IBD. Anti-TNF-alpha such as Infliximab (IFX), Adalimumab (ADA) and

Golimumab have proven to be effective in the treatment of IBD i.e. CD and UC as well as

(5)

in the treatment of rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis and

psoriasis. TNF-alpha antibodies are effective in inducing and maintaining remission

of luminal and fistulizing CD and UC.

1, 3, 4, 29

These drugs are administered at fixed dose

and intervals derived from dose finding studies for IFX and ADA.

30-33

Observational

studies showed that approximately 10% - 21% of the patients annually lose their

response to these TNF-alpha antibodies partially due to formation of antibodies against

the drug causing low drug trough levels.

34-36

The production of antidrug antibodies

is associated with infusion reactions and an accelerated antibody clearance resulting

in lower anti-TNF-alpha antibody titers.

37

Observational studies have demonstrated

a relationship between IFX and ADA drug concentrations, the presence of antidrug

antibodies and clinical outcome.

38-41

Strategies to prevent developing antibodies to

IFX and ADA are scheduled dosing to maintain stable trough drug levels and

co-administration of an immunomodulator (e.g. thiopurines). In the case of loss of

response with low drug titers without antibodies, increasing the dose or shortening of

the dosing interval is effective.

42

Whereas in cases of low drug titers due to anti-drug

antibody formation, a switch to another anti-TNF-alpha is the preferable strategy.

43

Therapeutic Drug and Immunogenicity Monitoring (TDIM) with early serial trough

and antidrug antibody level measurements will probably optimize anti-TNF-alpha

treatment.

44, 45

Therefore several studies are designed to dose IFX or ADA based on

trough levels. Different assays are being used to measure drug and anti-drug antibody

levels. The most commonly used assay types are the: enzyme-linked immunosorbent

assay (ELISA) and radio-immunoassay (RIA) and a fluid phase mobility shift assay.

Standardization of assays to measure IFX or ADA trough levels and IFX or

anti-ADA antibodies is lacking. Several confounding factors such as drug interference and

background can influence the measurement of biological drugs and antibodies to

these drugs. This may result in poor specificity, sensitivity and reproducibility. Our

aim in Chapter 5 was to determine the correlation between academically developed

assays (Leuven and Amsterdam) that were used in several studies to detect IFX drug

levels and anti-IFX antibodies

46-48

and a commercially available assay (further referred

to as BMD ELISA). The population pharmacokinetics of IFX have been described

earlier for patients with ankylosing spondylitis, rheumatoid arthritis and IBD.

49-51

However these models were not used to predict serum trough levels or for dose

optimizing of IFX. In this thesis we developed a pharmacokinetic (PK) model for

IFX in IBD patients for dose-optimization (Chapter 6). TNF-alpha blocking agents

are very expensive and currently constitute the majority of the costs of IBD therapy.

52

IFX-biosimilars, for example CT-P13 (Inflectra/ Remsima) and Flixabi (Biogen) are

available now.

53-55

These biosimilars are expected to have the same specificity and the

same sequence as the original molecule IFX (Remicade), but are marketed at a much

lower price than the first-generation anti-TNF-alpha blocking agents, which will

allow treatment of these diseases to be more cost-effective. Furthermore (Chapter

7) we want to determine whether antibodies to IFX (ATI) always neutralize the

anti-TNF-alpha drug and whether they show cross-reactivity towards other available

anti-TNF-alpha biologicals or IFX biosimilar. As described before some patients

treated with IFX or ADA develop ATI and this can result in loss of response. However,

(6)

Chapter

currently we are unable to predict or differentiate which patients are at risk of

developing these antibodies to IFX or ADA. One possible risk factor may be genetic

predisposition.

56, 57

As immunogenicity to anti-TNF alpha plays a role in the loss of

response to anti-TNF alpha therapy, our aim was to replicate known HLA regions and

identify novel (non-HLA) genetic regions associated with the development of anti

drug antibodies in patients with IBD (Chapter 8). IFX is administered intravenously.

This gives rise to certain disadvantages; patients need to visit the hospital for the

treatment, gets punctured and acute and late-onset infusion reactions could result

due to treatment.

58

Additionally, substantial side effects are expected as TNF-alpha is

an endogenous mediator and the patients receives IFX systemically causing systemic

immunosuppression.

59-61

Finally, ATI could develop leading to an increase in side

effects or loss of response to IFX therapy.

62, 63

These disadvantages have a negative

impact on health care cost and burden as well as patient-friendliness.

52

A part of these

disadvantages could be eliminated if IFX is administered orally, inducing a localized,

anti-inflammatory effect. Studies have shown that tissue IFX concentration correlates

with a better and sustained response in CD and that IFX exerts its effect at least partly

by local anti-inflammatory and immunomodulatory effects in the bowel.

64, 65

There are

uncontrolled pilot clinical trials that have shown that the local administration of IFX in

postoperative recurrent symptomatic isolated intestinal lesions and fistulising CD.

66-72

The ColoPulse technology is a coating technology which consist of a pH-sensitive

polymer in which a superdisintegrant is incorporated in the coating matrix.

73, 74

This

coating was specifically developed to target the ileo-colonic region in humans and is

characterized by fast and site-specific drug targeting. ColoPulse capsules and tablets

target the ileo-colonic region in healthy subjects as well as CD patients (Chapter 9).

75-79

Furthermore, we have shown that IFX compounded in ColoPulse tablets is feasible

and stable.

80

There is no published data on the efficacious dose of oral

ileo-colonic-targeted IFX. In Chapter 10 we propose a protocol Towards Mucosal Application of

IFX (TOMATE study). The objective of this protocol is to describe a study to treat

patients with active ileo-colonic CD with orally administered ColoPulse IFX tablets

instead of intravenously administrated IFX.

Outline of this thesis

Part I: mesalazine

In chapter 2 we describe symptoms of patients intolerant for 5-ASA after 5-ASA

rechallenge and thereafter the effectiveness of a rapid desensitization protocol in UC

patients with a demonstrated 5-ASA intolerance. In Chapter 3 we describe a patient

with therapy refractory UC who developed a mesalazine luxated auto-immune

hepatitis/PSC overlap syndrome.

In Chapter 4 we used a cohort who developed nephrotoxicity subsequent to

5-ASA administration to characterize the clinical features of this serious adverse event

and then perform the first genome wide association study to identify genetic risk

factors for the development of a drug-induced renal injury.

(7)

Part II: Anti-TNF alpha antibodies

In Chapter 5 we determined the correlation between academically developed assays

(Leuven and Amsterdam) that were used in several studies to detect IFX drug levels

and ATI and are routinely applied in patient diagnostics, and a commercially available

assay (further referred to as BMD ELISA). In Chapter 6 we compared the

TNF-alpha-neutralizing capacity of all commercially available anti-TNF-alpha drugs. Furthermore,

we tested the neutralizing capacity of ATI, as well as their cross-reactivity with IFX,

ATI, ADA and antibodies to Adalimumab. Finally, we tested if antibodies towards the

original IFX cross-react with the biosimilar of IFX (CT-P13 [Inflectra]). Our aim

in Chapter 7 was to replicate known HLA regions and identify novel (non-HLA)

genetic regions associated with the development of antibodies to IFX and ADA. In

Chapter 8 we described a retrospective study with pharmacokinetic model for IFX

in IBD patients that can be used for dose-optimization of IFX and to predict serum

trough levels in this population. In Chapter 9 we described that Colopulse tablets

perform comparably in healthy volunteers and CD patients and show no influence of

food and time of food intake on bioavailability. This chapter is a precursor study of

the protocol described in Chapter 10. In Chapter 10 we prepare a protocol Towards

Mucosal Application of IFX (TOMATE study). The objective of this protocol is to

describe a study to treat patients with active ileo-colonic CD with orally administered

ColoPulse IFX tablets instead of intravenously administrated IFX. Efficacy and safety

will be investigated as well as oral IFX pharmacokinetics and the development of ATI

due to oral IFX treatment.

(8)

Chapter

References

1. Van Assche G, Dignass A, Reinisch W, et al. The second European evidence-based

Consensus on the diagnosis and management of Crohn’s disease: Special situations. J Crohns

Colitis. 2010;4(1):63-101. doi: 10.1016/j.crohns.2009.09.009.

2. Spekhorst LM, Imhann F, Festen EAM, et al. Cohort profile: design and first results of the Dutch IBD Biobank: a prospective, nationwide biobank of patients with inflammatory bowel disease. BMJ Open. 2017;7(11):e016695-2017-016695. doi: 10.1136/

bmjopen-2017-016695 [doi].

3. Harbord M, Eliakim R, Bettenworth D, et al. Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 2: Current Management. J Crohns

Colitis. 2017;11(7):769-784. doi: 10.1093/ecco-jcc/jjx009 [doi].

4. Gomollon F, Dignass A, Annese V, et al. 3rd European Evidence-based Consensus on the Diagnosis and Management of Crohn’s Disease 2016: Part 1: Diagnosis and Medical Management. J Crohns Colitis. 2017;11(1):3-25. doi: 10.1093/ecco-jcc/jjw168 [doi]. 5. Ford AC, Achkar JP, Khan KJ, et al. Efficacy of 5-aminosalicylates in ulcerative colitis:

systematic review and meta-analysis. Am J Gastroenterol. 2011;106(4):601-616. doi: 10.1038/ajg.2011.67.

6. Loftus EV,Jr., Kane SV, Bjorkman D. Systematic review: short-term adverse effects of 5-aminosalicylic acid agents in the treatment of ulcerative colitis. Aliment Pharmacol Ther. 2004;19(0269-2813; 0269-2813; 2):179-189.

7. Flourie B, Hagege H, Tucat G, et al. Randomised clinical trial: once- vs. twice-daily prolonged-release mesalazine for active ulcerative colitis. Aliment Pharmacol Ther. 2013;37(8):767-775. doi: 10.1111/apt.12266 [doi].

8. Wang Y, Parker CE, Bhanji T, Feagan BG, MacDonald JK. Oral 5-aminosalicylic acid for induction of remission in ulcerative colitis. Cochrane Database Syst Rev. 2016;4:CD000543. doi: 10.1002/14651858.CD000543.pub4 [doi].

9. Feagan BG, Chande N, MacDonald JK. Are there any differences in the efficacy and safety of different formulations of Oral 5-ASA used for induction and maintenance of remission in ulcerative colitis? evidence from cochrane reviews. Inflamm Bowel Dis. 2013;19(9):2031-2040. doi: 10.1097/MIB.0b013e3182920108 [doi].

10. Di Paolo MC, Paoluzi OA, Pica R, et al. Sulphasalazine and 5-aminosalicylic acid in long-term treatment of ulcerative colitis: report on tolerance and side-effects. Dig Liver Dis. 2001;33(1590-8658; 1590-8658; 7):563-569.

11. Castells MC, Tennant NM, Sloane DE, et al. Hypersensitivity reactions to chemotherapy: outcomes and safety of rapid desensitization in 413 cases. J Allergy Clin Immunol. 2008;122(1097-6825; 1097-6825; 3):574-580.

12. Tolia V. Sulfasalazine desensitization in children and adolescents with chronic

inflammatory bowel disease. Am J Gastroenterol. 1992;87(0002-9270; 0002-9270; 8):1029-1032.

13. Paraskevopoulos I, Konstantinou G, Liatsos C. Desensitization treatment of an aspirin- and mesalamine-sensitive patient with Crohn’s disease. Inflamm Bowel Dis. 2005;11(1078-0998; 1078-0998; 4):417-418.

14. Stelzle RC, Squire EN. Oral desensitization to 5-aminosalicylic acid medications. Ann Allergy

Asthma Immunol. 1999;83(1081-1206; 1081-1206; 1):23-24.

15. Gonzalo MA, Alcalde MM, Garcia JM, Alvarado MI, Fernandez L. Desensitization after fever induced by mesalazine. Allergy. 1999;54(0105-4538; 0105-4538; 11):1224-1225. 16. Turunen U, Elomaa I, Anttila VJ, Seppala K. Mesalazine tolerance in patients with

inflammatory bowel disease and previous intolerance or allergy to sulphasalazine or sulphonamides. Scand J Gastroenterol. 1987;22(0036-5521; 0036-5521; 7):798-802. 17. Varela S, Diez MS, Gonzalez C, et al. Oral desensitization to 5-ASA. Allergy.

(9)

18. Oustamanolakis P, Koutroubakis IE. New desensitization regimen with mesalamine granules in a patient with ulcerative colitis and mesalamine intolerance. Inflamm Bowel Dis. 2010(1536-4844; 1078-0998).

19. Foster RA, Zander DS, Mergo PJ, Valentine JF. Mesalamine-related lung disease: clinical, radiographic, and pathologic manifestations. Inflamm Bowel Dis. 2003;9(0998; 1078-0998; 5):308-315.

20. Ransford RA, Langman MJ. Sulphasalazine and mesalazine: serious adverse reactions re-evaluated on the basis of suspected adverse reaction reports to the Committee on Safety of Medicines. Gut. 2002;51(0017-5749; 0017-5749; 4):536-539.

21. Van Staa TP, Travis S, Leufkens HG, Logan RF. 5-aminosalicylic acids and the risk of renal disease: a large British epidemiologic study. Gastroenterology. 2004;126(5085; 0016-5085; 7):1733-1739.

22. Stein RB, Hanauer SB. Comparative tolerability of treatments for inflammatory bowel disease. Drug Saf. 2000;23(5):429-448.

23. Gisbert JP, Gonzalez-Lama Y, Mate J. Thiopurine-induced liver injury in patients with inflammatory bowel disease: a systematic review. Am J Gastroenterol. 2007;102(7):1518-1527. doi: 10.1111/j.1572-0241.2007.01187.x.

24. Khokhar OS, Lewis JH. Hepatotoxicity of agents used in the management of inflammatory bowel disease. Dig Dis. 2010;28(3):508-518. doi: 10.1159/000320410.

25. Chester AC, Diamond LH, Schreiner GE. Hypersensitivity to salicylazosulfapyridine: renal and hepatic toxic reactions. Arch Intern Med. 1978;138(7):1138-1139.

26. Dwarakanath AD, Michael J, Allan RN. Sulphasalazine induced renal failure. Gut. 1992;33(7):1006-1007. doi: 10.1136/gut.33.7.1006 [doi].

27. Muller AF, Stevens PE, McIntyre AS, Ellison H, Logan RF. Experience of 5-aminosalicylate nephrotoxicity in the United Kingdom. Aliment Pharmacol Ther. 2005;21(10):1217-1224. doi: APT2462 [pii].

28. Gisbert JP, Luna M, Gonzalez-Lama Y, et al. Effect of 5-aminosalicylates on renal function in patients with inflammatory bowel disease: 4-year follow-up study. Gastroenterol Hepatol. 2008;31(8):477-484. doi: 13127088 [pii].

29. Stange EF, Travis SP, Vermeire S, et al. European evidence-based Consensus on the diagnosis and management of ulcerative colitis: Definitions and diagnosis. J Crohns Colitis. 2008;2(1):1-23. doi: 10.1016/j.crohns.2007.11.001.

30. Targan SR, Hanauer SB, van Deventer SJ, et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn’s Disease cA2 Study Group. N Engl J Med. 1997;337(15):1029-1035. doi: 10.1056/ NEJM199710093371502.

31. Rutgeerts P, Sandborn WJ, Feagan BG, et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2005;353(23):2462-2476. doi: 10.1056/ NEJMoa050516.

32. Hanauer SB, Sandborn WJ, Rutgeerts P, et al. Human anti-tumor necrosis factor

monoclonal antibody (adalimumab) in Crohn’s disease: the CLASSIC-I trial. Gastroenterology. 2006;130(2):323-33; quiz 591. doi: 10.1053/j.gastro.2005.11.030.

33. Sandborn WJ, van Assche G, Reinisch W, et al. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2012;142(2):257-65.e1-3. doi: 10.1053/j.gastro.2011.10.032.

34. Schnitzler F, Fidder H, Ferrante M, et al. Long-term outcome of treatment with infliximab in 614 patients with Crohn’s disease: results from a single-centre cohort. Gut. 2009;58(4):492-500. doi: 10.1136/gut.2008.155812.

35. Ainsworth MA, Bendtzen K, Brynskov J. Tumor necrosis factor-alpha binding capacity and anti-infliximab antibodies measured by fluid-phase radioimmunoassays as predictors of clinical efficacy of infliximab in Crohn’s disease. Am J Gastroenterol. 2008;103(4):944-948. doi: 10.1111/j.1572-0241.2007.01638.x.

(10)

Chapter

36. Qiu Y, Chen BL, Mao R, et al. Systematic review with meta-analysis: loss of response and requirement of anti-TNFalpha dose intensification in Crohn’s disease. J Gastroenterol. 2017;52(5):535-554. doi: 10.1007/s00535-017-1324-3 [doi].

37. Danese S, Fiorino G, Reinisch W. Review article: Causative factors and the clinical management of patients with Crohn’s disease who lose response to anti-TNF-alpha therapy. Aliment Pharmacol Ther. 2011;34(1):1-10. doi: 10.1111/j.1365-2036.2011.04679.x; 10.1111/j.1365-2036.2011.04679.x.

38. Maser EA, Villela R, Silverberg MS, Greenberg GR. Association of trough serum infliximab to clinical outcome after scheduled maintenance treatment for Crohn’s disease. Clin

Gastroenterol Hepatol. 2006;4(10):1248-1254. doi: 10.1016/j.cgh.2006.06.025.

39. Seow CH, Newman A, Irwin SP, Steinhart AH, Silverberg MS, Greenberg GR. Trough serum infliximab: a predictive factor of clinical outcome for infliximab treatment in acute ulcerative colitis. Gut. 2010;59(1):49-54. doi: 10.1136/gut.2009.183095.

40. Karmiris K, Paintaud G, Noman M, et al. Influence of trough serum levels and immunogenicity on long-term outcome of adalimumab therapy in Crohn’s disease.

Gastroenterology. 2009;137(5):1628-1640. doi: 10.1053/j.gastro.2009.07.062.

41. Billioud V, Sandborn WJ, Peyrin-Biroulet L. Loss of response and need for adalimumab dose intensification in Crohn’s disease: a systematic review. Am J Gastroenterol.

2011;106(4):674-684. doi: 10.1038/ajg.2011.60.

42. Katz L, Gisbert JP, Manoogian B, et al. Doubling the infliximab dose versus halving the infusion intervals in Crohn’s disease patients with loss of response. Inflamm Bowel Dis. 2012. doi: 10.1002/ibd.22902; 10.1002/ibd.22902.

43. Allez M, Karmiris K, Louis E, et al. Report of the ECCO pathogenesis workshop on anti-TNF therapy failures in inflammatory bowel diseases: definitions, frequency and pharmacological aspects. J Crohns Colitis. 2010;4(4):355-366. doi: 10.1016/j. crohns.2010.04.004 [doi].

44. Vande Casteele N, Ballet V, Van Assche G, Rutgeerts P, Vermeire S, Gils A. Early serial trough and antidrug antibody level measurements predict clinical outcome of infliximab and adalimumab treatment. Gut. 2012;61(2):321; author reply 322. doi: 10.1136/ gut.2010.236869.

45. van de Casteele N. 1159 Results on the Optimisation Phase of the Prospective Controlled Trough Level Adapted Infliximab Treatment (TAXIT) Trial. 5. 2012;142:211-212. 46. Jamnitski A, Bartelds GM, Nurmohamed MT, et al. The presence or absence of antibodies

to infliximab or adalimumab determines the outcome of switching to etanercept. Ann

Rheum Dis. 2011;70(2):284-288. doi: 10.1136/ard.2010.135111.

47. van Kuijk AW, de Groot M, Stapel SO, Dijkmans BA, Wolbink GJ, Tak PP. Relationship between the clinical response to adalimumab treatment and serum levels of adalimumab and anti-adalimumab antibodies in patients with psoriatic arthritis. Ann Rheum Dis. 2010;69(3):624-625. doi: 10.1136/ard.2009.108787.

48. Van Assche G, Vermeire S, Ballet V, et al. Switch to adalimumab in patients with Crohn’s disease controlled by maintenance infliximab: prospective randomised SWITCH trial. Gut. 2012;61(2):229-234. doi: 10.1136/gutjnl-2011-300755.

49. Ternant D, Aubourg A, Magdelaine-Beuzelin C, et al. Infliximab pharmacokinetics in inflammatory bowel disease patients. Ther Drug Monit. 2008;30(4):523-529. doi: 10.1097/ FTD.0b013e318180e300.

50. Fasanmade AA, Adedokun OJ, Ford J, et al. Population pharmacokinetic analysis of infliximab in patients with ulcerative colitis. Eur J Clin Pharmacol. 2009;65(12):1211-1228. doi: 10.1007/s00228-009-0718-4.

51. Fasanmade AA, Adedokun OJ, Blank M, Zhou H, Davis HM. Pharmacokinetic properties of infliximab in children and adults with Crohn’s disease: a retrospective analysis of data from 2 phase III clinical trials. Clin Ther. 2011;33(7):946-964. doi: 10.1016/j. clinthera.2011.06.002.

(11)

52. van der Valk ME, Mangen MJ, Leenders M, et al. Healthcare costs of inflammatory bowel disease have shifted from hospitalisation and surgery towards anti-TNFalpha therapy: results from the COIN study. Gut. 2014;63(1):72-79. doi: 10.1136/gutjnl-2012-303376 [doi].

53. Ebbers HC, Crow SA, Vulto AG, Schellekens H. Interchangeability, immunogenicity and biosimilars. Nat Biotechnol. 2012;30(12):1186-1190. doi: 10.1038/nbt.2438 [doi]. 54. Rinaudo-Gaujous M, Paul S, Tedesco ED, Genin C, Roblin X, Peyrin-Biroulet L. Review

article: biosimilars are the next generation of drugs for liver and gastrointestinal diseases.

Aliment Pharmacol Ther. 2013;38(8):914-924. doi: 10.1111/apt.12477 [doi].

55. Hong J, Lee Y, Lee C, et al. Physicochemical and biological characterization of SB2, a biosimilar of Remicade(R) (infliximab). MAbs. 2016:1-19. doi: 10.1080/19420862.2016.1264550 [doi].

56. Billiet T, Vande Casteele N, Van Stappen T, et al. Immunogenicity to infliximab is associated with HLA-DRB1. Gut. 2015;64(8):1344-1345. doi: 10.1136/gutjnl-2015-309698 [doi]. 57. Sazonovs A, Kennedy NA, Bewshea C, et al. HLA-DQA1 contributes to the development of

antibodies to anti-TNF therapy in Crohn’s disease. Oral presentation ECCO 2018. 58. Lichtenstein L, Ron Y, Kivity S, et al. Infliximab-Related Infusion Reactions: Systematic

Review. J Crohns Colitis. 2015;9(9):806-815. doi: 10.1093/ecco-jcc/jjv096 [doi]. 59. Shah ED, Farida JP, Siegel CA, Chong K, Melmed GY. Risk for Overall Infection with

Anti-TNF and Anti-integrin Agents Used in IBD: A Systematic Review and Meta-analysis. Inflamm

Bowel Dis. 2017;23(4):570-577. doi: 10.1097/MIB.0000000000001049 [doi].

60. Bonovas S, Fiorino G, Allocca M, et al. Biologic Therapies and Risk of Infection and Malignancy in Patients With Inflammatory Bowel Disease: A Systematic Review and Network Meta-analysis. Clin Gastroenterol Hepatol. 2016;14(10):1385-1397.e10. doi: 10.1016/j.cgh.2016.04.039 [doi].

61. Holleran G, Lopetuso L, Petito V, et al. The Innate and Adaptive Immune System as Targets for Biologic Therapies in Inflammatory Bowel Disease. Int J Mol Sci. 2017;18(10):10.3390/ ijms18102020. doi: E2020 [pii].

62. O’Meara S, Nanda KS, Moss AC. Antibodies to infliximab and risk of infusion reactions in patients with inflammatory bowel disease: a systematic review and meta-analysis. Inflamm

Bowel Dis. 2014;20(1):1-6. doi: 10.1097/01.MIB.0000436951.80898.6d [doi].

63. Nanda KS, Cheifetz AS, Moss AC. Impact of antibodies to infliximab on clinical outcomes and serum infliximab levels in patients with inflammatory bowel disease (IBD): a meta-analysis. Am J Gastroenterol. 2013;108(1):40-7; quiz 48. doi: 10.1038/ajg.2012.363 [doi]. 64. Yarur AJ, Jain A, Sussman DA, et al. The association of tissue anti-TNF drug levels with

serological and endoscopic disease activity in inflammatory bowel disease: the ATLAS study. Gut. 2016;65(2):249-255. doi: 10.1136/gutjnl-2014-308099 [doi].

65. Cornillie F, Shealy D, D’Haens G, et al. Infliximab induces potent anti-inflammatory and local immunomodulatory activity but no systemic immune suppression in patients with Crohn’s disease. Aliment Pharmacol Ther. 2001;15(4):463-473. doi: apt956 [pii].

66. Biancone L, Cretella M, Tosti C, et al. Local injection of infliximab in the postoperative recurrence of Crohn’s disease. Gastrointest Endosc. 2006;63(3):486-492. doi: S0016-5107(05)02756-2 [pii].

67. Lorenzo-Zuniga V, Boix J, Manosa M, et al. Local injection of infliximab in symptomatic isolated mucosal lesions: a novel scenario for endoscopic therapy?. Inflamm Bowel Dis. 2013;19(4):E59-61. doi: 10.1002/ibd.23018 [doi].

68. Alessandroni L, Kohn A, Cosintino R, et al. Local injection of infliximab in severe fistulating perianal Crohn’s disease: an open uncontrolled study. Tech Coloproctol. 2011;15(4):407-412. doi: 10.1007/s10151-011-0759-4 [doi].

69. Asteria CR, Ficari F, Bagnoli S, Milla M, Tonelli F. Treatment of perianal fistulas in Crohn’s disease by local injection of antibody to TNF-alpha accounts for a favourable clinical response in selected cases: a pilot study. Scand J Gastroenterol. 2006;41(9):1064-1072. doi: P351W04T13N14005 [pii].

(12)

Chapter

70. Poggioli G, Laureti S, Pierangeli F, et al. Local injection of adalimumab for perianal Crohn’s disease: better than infliximab?. Inflamm Bowel Dis. 2010;16(10):1631. doi: 10.1002/ibd.21210 [doi].

71. Hendel J, Karstensen JG, Vilmann P. Serial intralesional injections of infliximab in small bowel Crohn’s strictures are feasible and might lower inflammation. United European

Gastroenterol J. 2014;2(5):406-412. doi: 10.1177/2050640614547805 [doi].

72. Swaminath A, Lichtiger S. Dilation of colonic strictures by intralesional injection of infliximab in patients with Crohn’s colitis. Inflamm Bowel Dis. 2008;14(2):213-216. doi: 10.1002/ibd.20318 [doi].

73. Schellekens RC, Baltink JH, Woesthuis EM, et al. Film coated tablets (ColoPulse technology) for targeted delivery in the lower intestinal tract: influence of the core composition on release characteristics. Pharm Dev Technol. 2012;17(1):40-47. doi: 10.3109/10837450.2010.513986 [doi].

74. Schellekens RC, Stellaard F, Mitrovic D, Stuurman FE, Kosterink JG, Frijlink HW. Pulsatile drug delivery to ileo-colonic segments by structured incorporation of disintegrants in pH-responsive polymer coatings. J Control Release. 2008;132(2):91-98. doi: 10.1016/j. jconrel.2008.08.008 [doi].

75. Maurer JM, Schellekens RC, van Rieke HM, et al. Gastrointestinal pH and Transit Time Profiling in Healthy Volunteers Using the IntelliCap System Confirms Ileo-Colonic Release of ColoPulse Tablets. PLoS One. 2015;10(7):e0129076. doi: 10.1371/journal. pone.0129076 [doi].

76. Maurer JM, Schellekens RC, van Rieke HM, et al. ColoPulse tablets perform comparably in healthy volunteers and Crohn’s patients and show no influence of food and time of food intake on bioavailability. J Control Release. 2013;172(3):618-624. doi: 10.1016/j. jconrel.2013.09.021 [doi].

77. Maurer MJ, Schellekens RC, Wutzke KD, et al. A non-invasive, low-cost study design to determine the release profile of colon drug delivery systems: a feasibility study. Pharm Res. 2012;29(8):2070-2078. doi: 10.1007/s11095-012-0735-3 [doi].

78. Schellekens RC, Stellaard F, Olsder GG, Woerdenbag HJ, Frijlink HW, Kosterink JG. Oral ileocolonic drug delivery by the colopulse-system: a bioavailability study in healthy volunteers. J Control Release. 2010;146(3):334-340. doi: 10.1016/j.jconrel.2010.05.028 [doi].

79. Schellekens RC, Olsder GG, Langenberg SM, et al. Proof-of-concept study on the suitability of 13C-urea as a marker substance for assessment of in vivo behaviour of oral colon-targeted dosage forms. Br J Pharmacol. 2009;158(2):532-540. doi: 10.1111/j.1476-5381.2009.00302.x [doi].

80. Maurer JM, Hofman S, Schellekens RC, et al. Development and potential application of an oral ColoPulse infliximab tablet with colon specific release: A feasibility study. Int J Pharm. 2016;505(1-2):175-186. doi: 10.1016/j.ijpharm.2016.03.027 [doi].

(13)
(14)
(15)

Referenties

GERELATEERDE DOCUMENTEN

Different concentrations of IFX, IFX biosimilar CT-P13 (Inflectra), ADA, Certolizumab, Etanercept and Golimumab (0-5-10-20-40-100 ng/mL) were analyzed for their neutralizing

is correlated with the HLA-DRB1*03 allele. The HLA-DQA1*05 signal could thus be driving the HLA-DRB1*03 association signal. This highlights the need for additional studies to

Our simulations also showed that for patients without ATI, it may be considered to dose every 12 weeks instead of every 8 weeks: at dose relative to 400 mg / 8 weeks, this is

Bioavailability, lag- and pulse time were evaluated to determine whether there is any difference between healthy volunteers and Crohn’s patients in the in vivo drug

Primary objective of the study is to investigate the efficacy oral IFX biosimilar CT- P13 in ColoPulse tablets to induce clinical remission based on CDAI score of < 150 at week

A novel formulation of CT-P13 (infliximab biosimilar) for subcutaneous administration: 1-year result from a phase 1 open-label randomised controlled trial in patients with

Antistoffen gericht tegen TNF alpha (anti-TNF-alpha) zoals Infliximab en Adalimumab worden bij zowel de ziekte van Crohn als colitis ulcerosa gebruikt.. Tegen deze in de

Increasing Infliximab Dose Based on Symptoms, Biomarkers, and Serum Drug Concentrations Does Not Increase Clinical, Endoscopic, and Corticosteroid-Free Remission in Patients With