• No results found

Antibodies against antibodies: immunogenicity of adalimumab as a model - Chapter 1: General introduction

N/A
N/A
Protected

Academic year: 2021

Share "Antibodies against antibodies: immunogenicity of adalimumab as a model - Chapter 1: General introduction"

Copied!
20
0
0

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

Hele tekst

(1)

s

Antibodies against antibodies: immunogenicity of adalimumab as a model

van Schouwenburg, P.A.

Publication date

2012

Link to publication

Citation for published version (APA):

van Schouwenburg, P. A. (2012). Antibodies against antibodies: immunogenicity of

adalimumab as a model.

General rights

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), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

1

(3)

Chap

ter 1

In the last decade there have been major developments in the field of therapeutic monoclonal antibodies. At this point over thirty different therapeutic monoclonal antibodies have been approved by the FDA. These therapeutics are used for the treatment of cancer, auto-immune diseases and cardiovascular diseases. The majority of the patients respond very well to the treatment, however in some patients the therapeutic gets recognized by the patients’ immune system. This leads to antibody formation against the therapeutic, which has been linked to clinical non-response in these patients. Recognition by the immune system

Our immune system protects us against the pathogens we are exposed to every day. It consists of the interplay between the innate and the adaptive immune system. The innate immune system is the first line of defense and consists of the complement system and broadly recognizing pattern recognition receptors on innate immune cells such as macrophages and neutrophils. If the innate immune system fails to clear the pathogen swiftly, the adaptive immune system is activated. Upon the first encounter with a pathogen, the adaptive immune system develops immunological memory against this specific pathogen allowing it to respond more rapidly upon a second round of antigen exposure. The adaptive immune system consists of B and T cells each expressing its own antigen specific receptor. T cells proliferate and differentiate in response to antigen presentation to the T cell receptor (TCR) by antigen presenting cells. B cells recognize pathogens directly via their B cell receptor (BCR) and proliferate, maturate and differentiate upon target binding. Moreover, activated B cells produce antibodies with the same specificity as their BCR leading to neutralization of the pathogen and facilitating its clearance.1

Immunoglobulins

To be able to respond to all different types of pathogens, B cells require a large variation in BCR’s. Every BCR contains two heavy and two light chains, both consisting of a constant and a variable domain. The heavy chain variable region consists of a single variable (V), diversity (D) and joining (J) segment, which are combined during V(D)J recombination2,3.

In total there are ~40 V segments, ~25 D segments and ~6 J segments which can be combined to form a great variety of heavy chains. The light chain can be formed by recombination of either the kappa gene, in which ~40 V segments can be combined with one of ~5 J regions, or the lambda encoding region consisting of ~30 V regions and ~4 J regions (figure 1).4

V(D)J recombination leads to an enormous variation between BCR’s. This diversity is even further increased by random nucleotide incorporation in the joining regions during recombination and by a process called somatic hyper mutation.5 Somatic hyper mutation

(4)

Antigen binding can not only induce affinity maturation of the BCR, but it can also initiate isotype switching. In naive cells all antigen receptors expressed by B cells are of the IgM and IgD isotype.7 However, after antigen recognition, rearrangement in the heavy

chain coding region can lead to the expression of IgG, IgGA or IgE. The class of antibody produced in an immune response depends of the type of antigen the body is exposed to and the microenvironment of the B cell. IgA is mainly produced to protect mucosal surfaces, while IgE is involved in anti-parasitic immune responses and allergy.8;9 The IgG

isotype consists of four subtypes numbered according to the frequency in peripheral blood. IgG1 is mainly produced against bacterial pathogens; IgG2 is often directed against polysaccharides on bacterial surfaces.10 IgG3 is made as a response against viruses and

IgG4 is produced after long term exposure to allergens.11;12

is initiated after antigen binding and is a process in which small changes in the form of single nucleotide exchanges, deletions and duplications are introduced in the variable region of the BCR. Due to these small changes a large variety of B cells is made, each recognizing the same antigen with different binding affinities. B cells recognizing the antigen with the highest affinity are selected for outgrowth and survival.6

Figure 1: V(D)J recombination. The great variation between antibodies is, among other mechanisms,

the result of V(D)J recombination. The two heavy chains consist of a variable (V), diversity (D) and joining (J) segments. Both light chains are formed by recombination of a V and a J segment.

(5)

Chap

ter 1

Effector functions of immunoglobulins

The production of antibodies can trigger several downstream effector mechanisms. Antibodies can activate complement by binding to the C1 complex via their Fc tails.13

Activation of the complement system ends in the formation of the membrane attack complex leading to lysis of the target cell.14 Next to that, complement factors can opsonize

pathogens facilitating phagocytosis, and induce the release of anaphylatoxins, thereby promoting inflammation. IgG1, IgG3 and to a lesser extent IgG2 are able to activate the complement system, in contrast to IgG4 which is unable to activate the complement system.

Antibodies can also bind to Fc receptors via their Fc tail leading to activation of macrophages, monocytes, myeloid cells and dendritic cells. Activation of these receptors can lead to phagocytosis, endocytosis, antibody mediated cellular toxicity and cytokine release.15 Binding of Fc receptors also plays a role in the clearance of immune complexes.16

IgG1 and IgG3 are able to bind Fc receptors. IgG2 and IgG4 can also bind, but with a lower affinity.17

IgG4 is considered an odd antibody, not only is it limited in its effector functions, but it is able to exchange half molecules in vivo, leading to bispecific antibodies. Because of their bispecificity IgG4 antibodies are unable to cross-link identical antigens resulting in the formation of small immune complexes.18-20 Even though in general the percentage of

IgG4 in serum is low, it has been described that in some immune responses IgG4 can be the main subclass produced.21;22

Therapeutic monoclonal antibodies

When the development of monoclonal antibodies evolved, soon these antibodies were also developed for therapeutic purposes. Because of their specific targeting and powerful effector mechanisms to remove unwanted agents, monoclonal antibodies are considered very good therapeutics. In 1986, the first FDA approved monoclonal antibody was OKT3 which was used to prevent transplant rejection.23 OKT3 was a mouse monoclonal antibody

and one of the major problems in the clinic was immunogenicity. All patients treated with OKT3 developed anti-drug antibodies (ADA) against the therapeutic and these antibodies were linked to loss of clinical response.24 It was assumed that this immunogenicity

could be reduced by making therapeutic monoclonal antibodies that resembled human antibodies more closely. Therefore, a lot of effort was taken to humanize therapeutic monoclonal antibodies.

Humanization of monoclonal antibodies

The first step in the humanization process of therapeutic monoclonal antibodies was the development of chimeric antibodies (figure 2) of which the constant domain is fully

(6)

human whereas the variable region is murine. One example of a chimeric therapeutic antibody is infliximab, an anti-TNF agent used for the treatment of auto-immune diseases such as Rheumatoid arthritis (RA) (figure 3). For infliximab, reported immunogenicity numbers vary between 8-61%.25-31 The development of ADA is linked to lower infliximab

levels and clinical non-response.26;28-30

Later, humanized antibodies were developed; here the murine framework sequences around the complementarity determining regions (CDR’s) are replaced with human sequences and only the CDR’s are murine. One example of humanized therapeutic monoclonal antibodies is natalizumab, a monoclonal antibody directed against α-4 integrin, which is used for the treatment of multiple sclerosis. Despite humanization of the antibody, immunogenicity against natalizumab is found in 4-11% of the treated patients.32-36

As techniques developed further, this enabled the production of fully human antibodies using phage display or humanized mice. Two examples of fully human antibodies are golimumab and adalimumab, both anti-TNF antibodies used for the treatment of auto-immune diseases (figure 3). The hope was that these fully human antibodies would not elicit an immune response in patients. However, for both golimumab and adalimumab, immunogenicity has been reported. Although little data are available for golimumab, ADA were detected in two studies in 6.5-8.0% of the treated patients.37;38 For adalimumab

immunogenicity was reported in 1-44% of adalimumab treated patients.39-42 As was

found with other therapeutic proteins, ADA production is linked to lower adalimumab serum levels and reduced clinical response.

Next to fully human monoclonal antibodies, the development of other types of therapeutic proteins is emerging, hoping to reduce immunogenicity and increase specificity. Examples are certolizumab, a PEGylated Fab fragment directed against TNF and

Figure 2: Humanization of monoclonal antibodies. The first monoclonal antibodies available

for treatment were murine antibodies. Immunogenicity was a major problem in the treatment with these antibodies, which led to the development of antibodies with increased homology with human antibodies. Chimeric antibodies have a human (gray) constant domain, and a murine (black) variable region. In humanized antibodies only the CDR regions of the antibody are of murine origin. Currently, also fully human monoclonal antibodies are available for treatment.

(7)

Chap

ter 1

etanercept, a fusion protein between TNF receptor II and the Fc part of an IgG antibody (figure 3). For both drugs ADA formation has been described. Against certolizumab, ADA have been found in 5.1-8.1% of the patients, and for etanercept reports vary between 0-16%.43-47

The production of ADA against therapeutic antibodies has not only been linked to reduced clinical efficacy, but also is associated with increased frequency of clinical side effects. For example, several recent papers suggest a possible link between immunogenicity and increased risk of tromboembolic events.48-50 Together this indicates

that immunogenicity of therapeutic monoclonal antibodies is of clinical importance. There is a large variation in the prevalence of immunogenicity as reported in different studies. This can be explained by differences between patients groups, co-medication, timing of sampling, length of follow-up and drug dosing. Also the different types of assays used for detection of ADA will greatly influence the results as further discussed below.

Figure 3: The five anti-TNF agents available for the treatment of rheumatoid arthritis. Infliximab

is a chimeric antibody of which the variable domain is murine (black), while the constant domain is human (gray). Adalimumab is a fully human antibody. Etanercept is a fusion protein between a human IgG Fc tail and the TNF receptor. Golimumab is also a fully human antibody and certolizumab is a humanized Fab fragment which is PEGylated (light gray).

Assessment of immunogenicity

Many different assays are available for the measurement of ADA.51-54 A commonly used

assay is a bridging ELISA. In this assay format specific ADA in the test sample crosslink the drug which is used both for coating and detection. Another assay often used for ADA detection is an antigen binding test (ABT).29;55;56 In both mentioned assays drug

interference is a major complicating factor in the detection of ADA against therapeutic antibodies. Often the presence of drug levels in patient sera leads to false negative results in these assays.55 In assays that are sensitive for drug interference ADA are only detected

in case the amount of ADA produced exceeds drug levels. Since most patients are continuously treated with therapeutic monoclonal antibodies, drug interference often leads to an underestimation of ADA production. Immunogenicity data should therefore

(8)

always be interpreted in context of the assay used for ADA measurement. It is important to realize the discrepancy between the ADA levels produced in the patients and the ADA detected by the different assays. Drug interference also complicates investigation of the kinetics of the immune response against therapeutic monoclonal antibodies. Using an assay less sensitive for drug interference would lead to more reliable results.

Factors influencing immunogenicity

Patient related factors

There are many different factors that are thought to influence the immunogenicity of therapeutic proteins.53;57-59 First, the genetic background of patients will make some

patients more prone than others to develop ADA. For anti-TNF agents it was shown that patients developing ADA against their first anti-TNF agent are more likely to develop ADA against the second anti-TNF agent, suggesting that there is an intrinsic factor in these patients contributing to the immune response against therapeutic proteins.53;60 Until

now, only a single paper on genetic variation and immunogenicity of anti-TNF agents was described. In this paper a correlation between IL-10 genotype and antibody production against adalimumab is shown, but a causal relation was not further investigated.61 It can

be expected that MHC restriction might also predispose some patients for ADA production as described below. Collecting data on patient genetics and ADA formation is difficult due to large patient numbers required for this type of research to reach statistical power.

In addition, the immune status of the patient is thought to influence ADA formation. Patients with a highly active immune system are thought to have an increased risk for ADA development. This is supported by various reports that ADA producing patients show higher baseline disease activity and elevated levels of inflammation markers.62;63 Treatment strategy

The treatment regimen is thought to influence ADA formation as well. It was reported that in case of infliximab, higher dosing reduces immunogenicity. 25 Furthermore, the route of

administration seems to be important, since intravenous (IV) administration is thought to be less immunogenic compared to intra muscular or subcutaneous (SC) treatment.64-67 In

contrast to this, a study comparing SC and IV administration of abatacept (a fusion protein between the IgG1 Fc tail and CTLA-4) showed a mild increase in immunogenicity in the IV treated patients.68 Other studies suggest that the risk of immunogenicity increases with

prolonged treatment.59

Also the use of co-medication with immunomodulators has been linked to reduced immunogenicity. For azathioprine, mercaptopurine, hydrocortisone, and methotrexate it was described that co-treatment is linked to lower frequency of ADA formation in either RA of Crohns disease (CD) patients treated with anti-TNF agents.25;26;56;69;70

(9)

Chap

ter 1

Product related factors

Product related factors can also influence the immunogenicity of a therapeutic protein. First, the presence of B or T cell epitopes in the therapeutic protein will increase their immunogenic potential, as will be discussed more extensively below. Amino acid sequence differences between the therapeutic and endogenous protein may increase immunogenicity. Structural alteration of the therapeutic protein, such as aggregation, has been described to increase the risk of an immune reaction.71-73 The same is true for small

particles present in therapeutics.71 These aggregates and particles may be introduced

during the production and purification process of the therapeutic protein. It has been suggested that glycosylation of the therapeutic protein can influence the immunogenic potential.74

Target related factors

Therapeutic antibodies form immune complexes (IC) with their targets in vivo. It appears that the type of IC formed between a therapeutic monoclonal antibody and the target protein influences ADA formation. The formation of large IC might enhance uptake of the therapeutic antibody by antigen presenting cells. Large IC can also directly crosslink BCR’s leading to T cell independent B cell activation. Aggregates in the therapeutic can resemble immune complexes and induce immune activation in a similar fashion.75 In

case of anti-TNF agents there are reports on IC formation between TNF and some of the known anti-TNF agents.76;77 These studies show that adalimumab and infliximab can

form large complexes with TNF of respectively 4,000 kDa and 14,000 kDa. In contrast, etanercept only forms small IC complexes with a maximal size of 300 kDa (consistent with a 1:2 TNF: etanercept complex). In concordance with these data, immunogenicity against etanercept was only reported in few studies whereas others found low or no levels of ADA.

T cell epitopes

After phagocytosis of therapeutic proteins by antigen presenting cells, the protein is digested into peptides which can be presented via MHC molecules. Once the peptide is loaded, MHC is transported to the cell surface, where it can bind to specific T cell receptors (TCR) and induce proliferation of antigen specific T cells. Depending on the peptides available on the therapeutic protein and the HLA type of the patient, antigen presentation is more or less efficient.

To investigate the role of T cell epitopes in the immune response against therapeutic proteins different approaches can be used. First, in silico methods are used to identify T cell epitopes by the prediction of binding affinities of peptides to common HLA types.78

(10)

T cell proliferation in vitro. Furthermore linkage studies can indicate whether certain HLA alleles are linked to ADA formation.

Notwithstanding the fact that various groups are setting up systems to investigate T cell epitopes, little published data are available on T cell epitopes in anti-TNF agents, One study identified a T cell epitope associated with the G1m1 allotype, which is able to induce T cell proliferation in healthy donors with a different allotype.79 In case of

adalimumab, which is of the G1m1 allotype, this would suggest that patients with a G1m1 allotype are less likely to produce ADA against adalimumab. This is contradicted by a study by Bartelds et al showing increased immunogenicity in patients with the G1m1 allotype.60

Theoretically, knowing which peptides of a therapeutic protein result in T cell proliferation gives the opportunity to modify the protein to reduce immunogenicity. However, in case of anti-TNF the removal of T cell epitopes might not be cost-effective, since many alternative drugs are available and patients can easily switch to a different anti-TNF agent. However, increasing the knowledge on T cell epitopes will help the further development of in silico prediction tools and pre-clinical in vitro proliferation assays. These tools could be very valuable in reducing immunogenicity in a next generation of therapeutic proteins.

B cell epitopes

Mapping the binding sites of ADA on therapeutic proteins will enable the identification of B cell epitopes involved in immunogenicity. For golimumab and certolizumab no data are available on the immunogenic epitopes. For infliximab, one study shows that the immunogenic region of infliximab lies within the F(ab’)2 region.80 Another study by

Kosmac et al confirmed these results and performed experiments using linear peptides of the F(ab’)2 region to identify the precise B cell epitope. Unfortunately they did not succeed in this, suggesting that the immunogenic epitope is only present on correctly folded infliximab.81

Until now there are no data available for adalimumab on the identification of B cell epitopes. However, different assays for the detection of ADA use adalimumab F(ab’)2 for detection, indicating that the immunogenic region lies within the F(ab’)2 region of adalimumab. For etanercept, ADA have been described to be non-neutralizing, but the exact binding epitope has not been investigated.43;82-85

Identification of the precise epitopes involved in ADA binding could potentially allow their elimination from the therapeutic protein or shielding of these epitope using PEG groups or HIS tags.86;87 Of course such alterations would only be possible when small

numbers of immunogenic epitopes are involved and they are located outside the antigen binding domain to avoid destruction of target binding.

(11)

Chap

ter 1

ADA: neutralizing or binding antibodies?

There are two possible mechanisms by which the formation of ADA against therapeutic monoclonal antibodies can influence treatment efficacy. First, neutralizing ADA can block the binding of the therapeutic to its target. Second, ADA may form immune complexes with the therapeutic antibody, leading to increased clearance and reduced half life of the therapeutic. It is currently unknown which of these mechanisms is most important in clinical non-response to therapeutic antibody treatment.

For infliximab, two different studies show that at least part of the antibodies formed against infliximab have neutralizing capacity.80;81 In case of adalimumab, no such data are

available yet.

The size of IC formed determines their fate. Large IC will activate complement and will be rapidly cleared in the liver whereas small IC are not efficiently cleared and stay in circulation. Rojas et al. published a study investigating IC formation of infliximab in cynomolgus monkeys.88 They injected these monkeys with infliximab and a labeled

polyclonal anti-infliximab serum obtained from immunized rhesus monkeys and studied the size and clearance of the in vivo formed IC between infliximab and anti-infliximab antibodies. They observed the formation of IC of two different sizes, the largest being 670 kDa in size. As expected, the larger IC were more rapidly cleared from the circulation. Another study followed the formation of IC in three infliximab treated patients during infusion.89 In two of these patients only small IC were formed, while in the last patient

large IC were found. This last patient also developed a severe infusion reaction. This suggests that the size of IC formed between ADA and infliximab varies between patients. For adalimumab no such data are available.

The treatment of rheumatoid arthritis patients with adalimumab

In this thesis we investigate the role of immunogenicity against adalimumab in the treatment of rheumatoid arthritis (RA) patients with this therapeutic monoclonal antibody. RA is a systemic inflammatory disease that affects approximately 1% of the western population.90 The disease is characterized by chronic synovitis which can result in

destruction of cartilage and bone, leading to functional disability, joint damage, decreased quality of life and shortened life expectancy.91 Part of the RA patients are treated with

adalimumab, and in these patients the immunogenicity has been extensively linked to lower serum drug levels and reduced clinical response.30;39

Scope of this thesis

A variety of studies have been focusing on the clinical aspects of immunogenicity of adalimumab, in this thesis we focus on the immunological aspects. The role of IgG4

(12)

in the immune response against adalimumab is elucidated in chapter 2. In chapter 3 a novel assay for the detection of ADA against adalimumab is described, in which ADA can be measured in the presence of high drug levels. In chapter 4 this assay is used for the measurement of ADA in a cohort of 99 adalimumab treated RA patients during three years of treatment. This gives a more complete overview of the kinetics of the anti-adalimumab response. Chapter 5 describes that all antibodies against adalimumab are neutralizing, providing for the first time a mechanism by which ADA formation leads to clinical non-response. In chapter 6 we describe the production of human monoclonal antibodies against adalimumab, which are derived from patient peripheral blood. We study their affinity and the effect of the different affinities in various assay formats.

(13)

Chap

ter 1

ReFeReNCes

1. Dempsey PW, Allison ME, Akkaraju S, Goodnow CC, Fearon DT. C3d of complement as a molecular adjuvant: bridging innate and acquired immunity. Science 1996; 271(5247):348-50. 2. Alt FW, Yancopoulos GD, Blackwell TK, Wood C, Thomas E, Boss M et al. Ordered rearrangement

of immunoglobulin heavy chain variable region segments. EMBO J 1984; 3(6):1209-19. 3. Early P, Huang H, Davis M, Calame K, Hood L. An immunoglobulin heavy chain variable

region gene is generated from three segments of DNA: VH, D and JH. 1980. J Immunol 2004; 173(11):6503-14.

4. Melchers F, ten BE, Seidl T, Kong XC, Yamagami T, Onishi K et al. Repertoire selection by pre-B-cell receptors and B-pre-B-cell receptors, and genetic control of B-pre-B-cell development from immature to mature B cells. Immunol Rev 2000; 175:33-46.

5. Weigert M, Perry R, Kelley D, Hunkapiller T, Schilling J, Hood L. The joining of V and J gene segments creates antibody diversity. Nature 1980; 283(5746):497-9.

6. Odegard VH, Schatz DG. Targeting of somatic hypermutation. Nat Rev Immunol 2006; 6(8):573-83.

7. Taylor PC, Peters AM, Paleolog E, Chapman PT, Elliott MJ, McCloskey R et al. Reduction of chemokine levels and leukocyte traffic to joints by tumor necrosis factor alpha blockade in patients with rheumatoid arthritis. Arthritis Rheum 2000; 43(1):38-47.

8. Macpherson AJ, Hunziker L, McCoy K, Lamarre A. IgA responses in the intestinal mucosa against pathogenic and non-pathogenic microorganisms. Microbes Infect 2001; 3(12):1021-35.

9. Yazdanbakhsh M, van den Biggelaar A, Maizels RM. Th2 responses without atopy: immunoregulation in chronic helminth infections and reduced allergic disease. Trends Immunol 2001; 22(7):372-7.

10. Kelly-Quintos C, Cavacini LA, Posner MR, Goldmann D, Pier GB. Characterization of the opsonic and protective activity against Staphylococcus aureus of fully human monoclonal antibodies specific for the bacterial surface polysaccharide poly-N-acetylglucosamine. Infect Immun 2006; 74(5):2742-50.

11. Beck OE. Distribution of virus antibody activity among human IgG subclasses. Clin Exp Immunol 1981; 43(3):626-32.

12. Aalberse RC, Stapel SO, Schuurman J, Rispens T. Immunoglobulin G4: an odd antibody. Clin Exp Allergy 2009; 39(4):469-77.

13. Tao MH, Canfield SM, Morrison SL. The differential ability of human IgG1 and IgG4 to activate complement is determined by the COOH-terminal sequence of the CH2 domain. J Exp Med 1991; 173(4):1025-8.

14. Nauta AJ, Roos A, Daha MR. A regulatory role for complement in innate immunity and autoimmunity. Int Arch Allergy Immunol 2004; 134(4):310-23.

15. Ravetch JV, Bolland S. IgG Fc receptors. Annu Rev Immunol 2001; 19:275-90.

16. Kurlander RJ, Ellison DM, Hall J. The blockade of Fc receptor-mediated clearance of immune complexes in vivo by a monoclonal antibody (2.4G2) directed against Fc receptors on murine leukocytes. J Immunol 1984; 133(2):855-62.

17. Nirula A, Glaser SM, Kalled SL, Taylor FR. What is IgG4? A review of the biology of a unique immunoglobulin subtype. Curr Opin Rheumatol 2011; 23(1):119-24.

18. Aalberse RC, Schuurman J, van Ree R. The apparent monovalency of human IgG4 is due to bispecificity. Int Arch Allergy Immunol 1999; 118(2-4):187-9.

(14)

antibodies form small, nonprecipitating immune complexes due to functional monovalency. J Immunol 1986; 137(11):3566-71.

20. van der Neut KM, Schuurman J, Losen M, Bleeker WK, Martinez-Martinez P, Vermeulen E et al. Anti-inflammatory activity of human IgG4 antibodies by dynamic Fab arm exchange. Science 2007; 317(5844):1554-7.

21. Aalberse RC, van der Gaag R, van Leeuwen J. Serologic aspects of IgG4 antibodies. I. Prolonged immunization results in an IgG4-restricted response. J Immunol 1983; 130(2):722-6.

22. van der Giessen M, Homan WL, van KG, Aalberse RC, Dieges PH. Subclass typing of IgG antibodies formed by grass pollen-allergic patients during immunotherapy. Int Arch Allergy Appl Immunol 1976; 50(5):625-40.

23. Prentice HG, Blacklock HA, Janossy G, Bradstock KF, Skeggs D, Goldstein G et al. Use of anti-T-cell monoclonal antibody OKT3 to prevent acute graft-versus-host disease in allogeneic bone-marrow transplantation for acute leukaemia. Lancet 1982; 1(8274):700-3.

24. Goldstein G, Fuccello AJ, Norman DJ, Shield CF, III, Colvin RB, Cosimi AB. OKT3 monoclonal antibody plasma levels during therapy and the subsequent development of host antibodies to OKT3. Transplantation 1986; 42(5):507-11.

25. Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, Macfarlane JD et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41(9):1552-63.

26. Baert F, Noman M, Vermeire S, Van Assche G, D’ Haens G, Carbonez A et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn’s disease. N Engl J Med 2003; 348(7):601-8.

27. Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld FC, Kalden JR et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med 2000; 343(22):1594-602.

28. Wolbink GJ, Vis M, Lems W, Voskuyl AE, de Groot E, Nurmohamed MT et al. Development of antiinfliximab antibodies and relationship to clinical response in patients with rheumatoid arthritis. Arthritis Rheum 2006; 54(3):711-5.

29. Bendtzen K, Geborek P, Svenson M, Larsson L, Kapetanovic MC, Saxne T. [Treatment of rheumatoid arthritis (RA) with anti-TNF-alpha antibody (Remicade). Individual monitoring of bioavailability and immunogenicity--secondary publication]. Ugeskr Laeger 2007; 169(5):420-3.

30. Radstake TR, Svenson M, Eijsbouts AM, van den Hoogen FH, Enevold C, van Riel PL et al. Formation of antibodies against infliximab and adalimumab strongly correlates with functional drug levels and clinical responses in rheumatoid arthritis. Ann Rheum Dis 2008;ard.

31. Finckh A, Ciurea A, Brulhart L, Kyburz D, Moller B, Dehler S et al. B cell depletion may be more effective than switching to an alternative anti-tumor necrosis factor agent in rheumatoid arthritis patients with inadequate response to anti-tumor necrosis factor agents. Arthritis Rheum 2007; 56(5):1417-23.

32. Calabresi PA, Giovannoni G, Confavreux C, Galetta SL, Havrdova E, Hutchinson M et al. The incidence and significance of anti-natalizumab antibodies: results from AFFIRM and SENTINEL. Neurology 2007; 69(14):1391-403.

33. Haghikia A, Fischer M, Hellwig K, Linker R, Chan A, Hohlfeld R et al. [Open use of natalizumab. Neutralising antibodies and clinical data]. Nervenarzt 2008; 79(6):716-9.

(15)

Chap

ter 1

35. Miller DH, Khan OA, Sheremata WA, Blumhardt LD, Rice GP, Libonati MA et al. A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2003; 348(1):15-23. 36. Oturai AB, Koch-Henriksen N, Petersen T, Jensen PE, Sellebjerg F, Sorensen PS. Efficacy of

natalizumab in multiple sclerosis patients with high disease activity: a Danish nationwide study. Eur J Neurol 2009; 16(3):420-3.

37. Zhou H, Jang H, Fleischmann RM, Bouman-Thio E, Xu Z, Marini JC et al. Pharmacokinetics and safety of golimumab, a fully human anti-TNF-alpha monoclonal antibody, in subjects with rheumatoid arthritis. J Clin Pharmacol 2007; 47(3):383-96.

38. Smolen J, Landewe RB, Mease P, Brzezicki J, Mason D, Luijtens K et al. Efficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the RAPID 2 study. A randomised controlled trial. Ann Rheum Dis 2009; 68(6):797-804.

39. Bartelds GM, Krieckaert CL, Nurmohamed MT, van Schouwenburg PA, Lems WF, Twisk JW et al. Development of antidrug antibodies against adalimumab and association with disease activity and treatment failure during long-term follow-up. JAMA 2011; 305(14):1460-8.

40. Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA et al. Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum 2003; 48(1):35-45.

41. van De Putte LB, Atkins C, Malaise M, Sany J, Russell AS, van Riel PL et al. Efficacy and safety of adalimumab as monotherapy in patients with rheumatoid arthritis for whom previous disease modifying antirheumatic drug treatment has failed. Ann Rheum Dis 2004; 63(5):508-16. 42. Miyasaka N. Clinical investigation in highly disease-affected rheumatoid arthritis patients in

Japan with adalimumab applying standard and general evaluation: the CHANGE study. Mod Rheumatol 2008; 18(3):252-62.

43. Anderson PJ. Tumor necrosis factor inhibitors: clinical implications of their different immunogenicity profiles. Semin Arthritis Rheum 2005; 34(5 Suppl1):19-22.

44. de Vries MK, van der Horst-Bruinsma IE, Nurmohamed MT, Aarden LA, Stapel SO, Peters MJ et al. Immunogenicity does not influence treatment with etanercept in patients with ankylosing spondylitis (AS). Ann Rheum Dis 2008.

45. Choy EH, Hazleman B, Smith M, Moss K, Lisi L, Scott DG et al. Efficacy of a novel PEGylated humanized anti-TNF fragment (CDP870) in patients with rheumatoid arthritis: a phase II double-blinded, randomized, dose-escalating trial. Rheumatology (Oxford) 2002; 41(10):1133-7.

46. Fleischmann R, Vencovsky J, van Vollenhoven RF, Borenstein D, Box J, Coteur G et al. Efficacy and safety of certolizumab pegol monotherapy every 4 weeks in patients with rheumatoid arthritis failing previous disease-modifying antirheumatic therapy: the FAST4WARD study. Ann Rheum Dis 2009; 68(6):805-11.

47. Jamnitski A, Krieckaert CL, Nurmohamed MT, Hart MH, Dijkmans BA, Aarden L et al. Patients non-responding to etanercept obtain lower etanercept concentrations compared with responding patients. Ann Rheum Dis 2012; 71(1):88-91.

48. Petitpain N, Gambier N, Wahl D, Chary-Valckenaere I, Loeuille D, Gillet P. Arterial and venous thromboembolic events during anti-TNF therapy: a study of 85 spontaneous reports in the period 2000-2006. Biomed Mater Eng 2009; 19(4-5):355-64.

49. Makol A, Grover M, Guggenheim C, Hassouna H. Etanercept and venous thromboembolism: a case series. J Med Case Reports 2010; 4:12.

50. Korswagen LA, Bartelds GM, Krieckaert CL, Turkstra F, Nurmohamed MT, van SD et al. Venous and arterial thromboembolic events in adalimumab-treated patients with antiadalimumab

(16)

antibodies: A case series and cohort study. Arthritis Rheum 2011; 63(4):877-83.

51. Aarden L, Ruuls SR, Wolbink G. Immunogenicity of anti-tumor necrosis factor antibodies-toward improved methods of anti-antibody measurement. Curr Opin Immunol 2008; 20(4):431-5.

52. Krieckaert C, Rispens T, Wolbink G. Immunogenicity of biological therapeutics: from assay to patient. Curr Opin Rheumatol 2012; 24(3):306-11.

53. Chirmule N, Jawa V, Meibohm B. Immunogenicity to Therapeutic Proteins: Impact on PK/PD and Efficacy. AAPS J 2012; 14(2):296-302.

54. Koren E, Smith HW, Shores E, Shankar G, Finco-Kent D, Rup B et al. Recommendations on risk-based strategies for detection and characterization of antibodies against biotechnology products. J Immunol Methods 2008; 333(1-2):1-9.

55. Hart MH, de VH, Wouters D, Wolbink GJ, Killestein J, de Groot ER et al. Differential effect of drug interference in immunogenicity assays. J Immunol Methods 2011; 372(1-2):196-203. 56. Bartelds GM, Wijbrandts CA, Nurmohamed MT, Stapel S, Lems WF, Aarden L et al. Clinical

response to adalimumab: relationship to anti-adalimumab antibodies and serum adalimumab concentrations in rheumatoid arthritis. Ann Rheum Dis 2007; 66(7):921-6.

57. Carter P. Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer 2001; 1(2):118-29.

58. Clark M. Antibody humanization: a case of the ‘Emperor’s new clothes’? Immunol Today 2000; 21(8):397-402.

59. Schellekens H. Immunogenicity of therapeutic proteins: clinical implications and future prospects. Clin Ther 2002; 24(11):1720-40.

60. Bartelds GM, de GE, Nurmohamed MT, Hart MH, van Eede PH, Wijbrandts CA et al. Surprising negative association between IgG1 allotype disparity and anti-adalimumab formation: a cohort study. Arthritis Res Ther 2010; 12(6):R221.

61. Bartelds GM, Wijbrandts CA, Nurmohamed MT, Wolbink GJ, de Vries N, Tak PP et al. Anti-adalimumab antibodies in rheumatoid arthritis patients are associated with interleukin-10 gene polymorphisms. Arthritis Rheum 2009; 60(8):2541-2.

62. Jamnitski A, Bartelds GM, Nurmohamed MT, van Schouwenburg PA, van SD, Stapel SO 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):284-8.

63. Bendtzen K, Geborek P, Svenson M, Larsson L, Kapetanovic MC, Saxne T. Individualized monitoring of drug bioavailability and immunogenicity in rheumatoid arthritis patients treated with the tumor necrosis factor alpha inhibitor infliximab. Arthritis Rheum 2006; 54(12):3782-9.

64. Schellekens H. Factors influencing the immunogenicity of therapeutic proteins. Nephrol Dial Transplant 2005; 20 Suppl 6:vi3-vi9.

65. Ross C, Clemmesen KM, Svenson M, Sorensen PS, Koch-Henriksen N, Skovgaard GL et al. Immunogenicity of interferon-beta in multiple sclerosis patients: influence of preparation, dosage, dose frequency, and route of administration. Danish Multiple Sclerosis Study Group. Ann Neurol 2000; 48(5):706-12.

66. Porter S. Human immune response to recombinant human proteins. J Pharm Sci 2001; 90(1):1-11.

67. Konrad MW, Childs AL, Merigan TC, Borden EC. Assessment of the antigenic response in humans to a recombinant mutant interferon beta. J Clin Immunol 1987; 7(5):365-75.

68. Genovese MC, Covarrubias A, Leon G, Mysler E, Keiserman M, Valente R et al. Subcutaneous abatacept versus intravenous abatacept: a phase IIIb noninferiority study in patients with an

(17)

Chap

ter 1

inadequate response to methotrexate. Arthritis Rheum 2011; 63(10):2854-64.

69. Farrell RJ, Alsahli M, Jeen YT, Falchuk KR, Peppercorn MA, Michetti P. Intravenous hydrocortisone premedication reduces antibodies to infliximab in Crohn’s disease: a randomized controlled trial. Gastroenterology 2003; 124(4):917-24.

70. Krieckaert CL, Bartelds GM, Lems WF, Wolbink GJ. The effect of immunomodulators on the immunogenicity of TNF-blocking therapeutic monoclonal antibodies: a review. Arthritis Res Ther 2010; 12(5):217.

71. Carpenter J, Cherney B, Lubinecki A, Ma S, Marszal E, Mire-Sluis A et al. Meeting report on protein particles and immunogenicity of therapeutic proteins: filling in the gaps in risk evaluation and mitigation. Biologicals 2010; 38(5):602-11.

72. Chirino AJ, Mire-Sluis A. Characterizing biological products and assessing comparability following manufacturing changes. Nat Biotechnol 2004; 22(11):1383-91.

73. Hermeling S, Schellekens H, Maas C, Gebbink MF, Crommelin DJ, Jiskoot W. Antibody response to aggregated human interferon alpha2b in wild-type and transgenic immune tolerant mice depends on type and level of aggregation. J Pharm Sci 2006; 95(5):1084-96.

74. Jefferis R. Glycosylation of recombinant antibody therapeutics. Biotechnol Prog 2005; 21(1):11-6.

75. Rosenberg AS. Effects of protein aggregates: an immunologic perspective. AAPS J 2006; 8(3):E501-E507.

76. Kohno T, Tam LT, Stevens SR, Louie JS. Binding characteristics of tumor necrosis factor receptor-Fc fusion proteins vs anti-tumor necrosis factor mAbs. J Investig Dermatol Symp Proc 2007; 12(1):5-8.

77. Kim MS, Lee SH, Song MY, Yoo TH, Lee BK, Kim YS. Comparative analyses of complex formation and binding sites between human tumor necrosis factor-alpha and its three antagonists elucidate their different neutralizing mechanisms. J Mol Biol 2007; 374(5):1374-88.

78. Brusic V, Bajic VB, Petrovsky N. Computational methods for prediction of T-cell epitopes--a framework for modelling, testing, and applications. Methods 2004; 34(4):436-43.

79. Stickler MM, Reddy A, Xiong JM, Hinton PR, DuBridge R, Harding FA. The human G1m1 allotype associates with CD4+ T-cell responsiveness to a highly conserved IgG1 constant region peptide and confers an asparaginyl endopeptidase cleavage site. Genes Immun 2011; 12(3):213-21. 80. Ben-Horin S, Yavzori M, Katz L, Kopylov U, Picard O, Fudim E et al. The immunogenic part of

infliximab is the F(ab’)2, but measuring antibodies to the intact infliximab molecule is more clinically useful. Gut 2010.

81. Kosmac M, Avcin T, Toplak N, Simonini G, Cimaz R, Curin S, V. Exploring the binding sites of anti-infliximab antibodies in pediatric patients with rheumatic diseases treated with anti-infliximab. Pediatr Res 2011; 69(3):243-8.

82. Lobo ED, Hansen RJ, Balthasar JP. Antibody pharmacokinetics and pharmacodynamics. J Pharm Sci 2004; 93(11):2645-68.

83. Dore RK, Mathews S, Schechtman J, Surbeck W, Mandel D, Patel A et al. The immunogenicity, safety, and efficacy of etanercept liquid administered once weekly in patients with rheumatoid arthritis. Clin Exp Rheumatol 2007; 25(1):40-6.

84. Moreland LW, Schiff MH, Baumgartner SW, Tindall EA, Fleischmann RM, Bulpitt KJ et al. Etanercept therapy in rheumatoid arthritis. A randomized, controlled trial. Ann Intern Med 1999; 130(6):478-86.

85. Weinblatt ME, Kremer JM, Bankhurst AD, Bulpitt KJ, Fleischmann RM, Fox RI et al. A trial of etanercept, a recombinant tumor necrosis factor receptor:Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate. N Engl J Med 1999; 340(4):253-9.

(18)

86. Mayer A, Sharma SK, Tolner B, Minton NP, Purdy D, Amlot P et al. Modifying an immunogenic epitope on a therapeutic protein: a step towards an improved system for antibody-directed enzyme prodrug therapy (ADEPT). Br J Cancer 2004; 90(12):2402-10.

87. Tsutsumi Y, Onda M, Nagata S, Lee B, Kreitman RJ, Pastan I. Site-specific chemical modification with polyethylene glycol of recombinant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) improves antitumor activity and reduces animal toxicity and immunogenicity. Proc Natl Acad Sci U S A 2000; 97(15):8548-53.

88. Rojas JR, Taylor RP, Cunningham MR, Rutkoski TJ, Vennarini J, Jang H et al. Formation, distribution, and elimination of infliximab and anti-infliximab immune complexes in cynomolgus monkeys. J Pharmacol Exp Ther 2005; 313(2):578-85.

89. van der Laken CJ, Voskuyl AE, Roos JC, Stigter vW, de Groot ER, Wolbink G et al. Imaging and serum analysis of immune complex formation of radiolabelled infliximab and anti-infliximab in responders and non-responders to therapy for rheumatoid arthritis. Ann Rheum Dis 2007; 66(2):253-6.

90. Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part II. Arthritis Rheum 2008; 58(1):26-35.

91. Symmons DP. Epidemiology of rheumatoid arthritis: determinants of onset, persistence and outcome. Best Pract Res Clin Rheumatol 2002; 16(5):707-22.

(19)
(20)

Referenties

GERELATEERDE DOCUMENTEN

De gedreven Groningse onderwijskundige, die zich ruim vijftien jaar sterk had gemaakt voor de middenschool, moest aan het einde van zijn loopbaan constateren dat de

Waar Nederlandse leraren en onderwijsbestuurders vreesden dat buitenlanders een bedreiging vormden voor de beginselen van individuele vrijheid, gelijkheid en

Het onderscheid tussen ‘witte’ en ‘zwarte’ scholen werd in deze tijd gemeengoed, de eerste islamitische scholen werden in de jaren tachtig opgericht,

Jongeren in Nederland in de jaren vijftig en zestig’, Bijdragen en Mededelingen betreffende de Geschiedenis der Nederlanden 101 (1986) 76-94.. Rovers, F., ‘Eenheid

This study seeks to trace the Dutch quest for individual freedom and autonomy historically, accounting for the development and the consequences of the growing faith

Door discussies over onderwijs en jeugd vanaf 1945 systematisch te bestuderen, ontstaat inzicht in de opkomst van een maatschappelijk zelfbeeld: het beeld van Nederland

We check the accuracy of our policy function using a grid of capital holdings that is much more dense than the one used to obtain the numerical solution and the grid points for

‘De uitzendovereenkomst is de arbeidsovereenkomst waarbij de werknemer door de werk- gever, in het kader van de uitoefening van het beroep of bedrijf van de werkgever dat is gericht