• No results found

B cells and B cell directed therapies in rheumatoid arthritis: towards personalized medicine - Chapter 12: General discussion

N/A
N/A
Protected

Academic year: 2021

Share "B cells and B cell directed therapies in rheumatoid arthritis: towards personalized medicine - Chapter 12: General discussion"

Copied!
7
0
0

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

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

B cells and B cell directed therapies in rheumatoid arthritis: towards

personalized medicine

Thurlings, R.M.

Publication date

2011

Link to publication

Citation for published version (APA):

Thurlings, R. M. (2011). B cells and B cell directed therapies in rheumatoid arthritis: towards

personalized medicine.

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)

PAGE. 78PAGE. 79 – Chapter 12

B Cells and B Cell directed therapies in Rheumatiod Arthritis

CHAPTER



GENERAL

DISCUSSION

B Cells and B Cell directed therapies in Rheumatiod Arthritis

(3)

PAGE. 80PAGE. 8 

gressive disease course and differential response to therapy 3. In line with this hypothesis the

presence of SLN was found to be associated with RF positive disease, the presence of erosions and rheumatoid nodules 4-6. However these

observa-tions were made in small patient cohorts using a selection of patients with end-stage disease. In this thesis we studied whether the presence of SLN can be used as a biomarker to identify clinically and immunological patient subsets and whether, as such, its presence can be used to predict clinical response to treatment.

In chapters 2 and 3 we analyzed the re-lationship between SLN, humoral autoimmunity, clinical disease characteristics and treatment re-sponse in a large and representative cohort of RA patients. First, we analyzed the relationship be-tween SLN and disease characteristics. We found that the presence of SLN is related to increased local and systemic inflammatory parameters, but it is found equally frequent in seropositive and seronegative RA patients. Furthermore, SLN was not associated with increased clinical disease activity parameters or the presence of erosive or nodular disease. We recently confirmed and extended these findings in a prospective cohort of patients with early arthritis 7. The presence of

SLN was neither associated with diagnosis nor with development of persistent or erosive disease. The presence of SLN decreased over time in indi-vidual patients, which probably reflected reduced synovial inflammation after anti-inflammatory therapy or self-limiting synovitis in this particular cohort.

The presence of SLN in both seronega-tive and seroposiseronega-tive RA ,as well as other forms of arthritis, suggests that SLN is a phenomenon which is not necessarily associated with autoan-tibodies. In line with this we recently found that even in seropositive patients the presence of SLN is not associated with an increase in local ACPA or RF production compared to other antibodies

8. Taken together, despite the fact that SLN has

germinal center-like features, the data imply that SLN is rather an inflammation-driven humoral response phenomenon than a true germinal center reaction that refines and amplifies a local autoantibody response. Still, SLN may contribute to inflammation by enhancing T cell activation, promoting local differentiation of plasma cells and by production of cytokines.

In chapter 3 we analyzed whether SLN predicts the response to TNF blockade. We found that patients with SLN overall responded well to TNF blockade and that SLN predicted clinical response independent of other known predictors, such as disease activity at baseline, the presence of ACPA and the level of synovial TNFα expres-sion. Furthermore, we found that SLN is rapidly reversible after treatment. SLN tends to disap-pear already 48 hours after treatment, before a decrease in DAS28 occurs. These data suggest that TNF is involved in the formation of SLN. In line with this, previous studies showed that TNF blockade results in decreased expression of cyto-kines 9, chemokines 10, and adhesion molecules 11,

which are required for secondary lymphoid organ formation.

In summary, we found that SLN does not define a clinically distinct disease phenotype. Furthermore, the data suggest that while SLN exhibits some features of germinal centers it does not function as such. As an inflammation driven humoral immune phenomenon it predicts a good response to infliximab. Nonetheless, the predic-tive value of SLN as a biomarker of response to TNF blockade in itself is not high enough to be used as a solitary biomarker, indicating the in-volvement of other, as yet unknown mechanisms. Future work should expand the search for other biomarkers and molecular networks, as well as combinations of clinical variables, to achieve an effective approach that will increase the percent-age of patients exhibiting a robust response to TNF blockade.

INTRODUCTION Within the past decade a num-ber of novel effective treatments have become available for rheumatoid arthritis (RA), among which tumor necrosis factor (TNF) blockers, rituximab, abatacept and tocizilumab. Nonethe-less, the response to these treatments differs be-tween RA patients and disease remission is only achieved in a proportion of patients and patients need to be treated chronically with often relatively expensive drugs. There is therefore a need to further improve the treatment of RA. This could be achieved by combination of different strate-gies. First, more optimal use of currently avail-able treatments is needed. The use of biomarkers could play a role in optimising therapy in individ-ual patients. For this purpose, biomarkers need to be identified that are predictive which patients will benefit most from a specific treatment. We also need to understand which immune mecha-nisms continue to drive the disease in patients who do not respond to current therapies. Sub-sequently, we can use this knowledge to develop novel therapies and to optimize current treatment schedules.

Multiple lines of evidence suggest RA is a het-erogeneous disease with a variable response to targeted therapy. As discussed in chapter 1 this heterogeneity is observed at the clinical, genetic and immunological level. Detailed analysis of im-munological mechanisms in RA and assessment of their relationship with genetic and environ-mental risk factors, clinical characteristics and treatment response may improve our understand-ing of the pathophysiologic heterogeneity in RA,

yield biomarkers to predict response and help to design novel treatments. In this thesis we focussed on B cells and B cell-directed therapies. We investigated whether we could identify bio-markers predictive of the clinical response to TNF blockade and rituximab treatment. Furthermore, we evaluated repeated treatment in initial clinical responders versus non-responders to rituximab. Finally, we performed a phase I study with the novel B cell modulating agent atacicept.

SYNOVIAL LYMPHOID NEOGENESIS AS A BIOMARKER FOR RESPONSE TO TNF BLOCKADE IN RA. The synovial tissue of RA patients is infiltrated predominantly by mac-rophages and lymphocytes, but also contains dendritic cells, mast cells, natural killer cells and neutrophilic granulocytes. When focussing on lymphocytes, the level of infiltration differs between patients. In some patients the tissue con-tains a diffuse or scarce infiltrate of T cells, while in others B and T cells are organized in lympho-cyte aggregates, which are surrounded by fields of plasma cells 1,2. Large lymphocyte aggregates may

exhibit features of germinal centers of second-ary lymphoid tissue, which has been termed synovial lymphoid neogenesis (SLN). This raises the question whether they refine and amplify local humoral autoimmune responses against for instance rheumatoid factor (RF) and anti-citrul-linated peptide antibodies (ACPA).

It has been proposed that synovial tissue with SLN forms a pathophysiologic subtype of RA, characterized by amplified local autoantibody production, associated with a more severe,

ag-GENERAL

DISCUSSION

Chapter 12

(4)

PAGE. 8 PAGE. 8 

and/or ACPA. Previous work has shown that au-toantibody positive RA patients respond better to rituximab treatment than those who are autoan-tibody negative 27. Taken together, these data

sup-port the notion that autoantibody production by B cell-derived plasma cells plays an important role in promoting synovial inflammation. Conceivably, effectiveness of rituximab treatment may be in-creased in autoantibody positive non-responders by strategies aimed at suppression of plasma cells associated with autoimmunity in the tissues.

In chapter 7 we studied the relationship between the response to rituximab and the type I interferon (IFN) signature in peripheral blood mononuclear cells (PBMCs). The type I IFN sig-nature is found in a subset of RA patients as the dominant signalling signature in PBMCs. It is also found in a proportion of patients with other auto-antibody associated autoimmune diseases, such as Sjögren’s syndrome, multiple sclerosis, der-matomyositis, type I diabetes mellitus, systemic sclerosis and systemic lupus erythematosus 28-33.

Rituximab has shown to be clinically efficacious in many of these conditions 34;35. In the two cohorts

that we studied rituximab was less effective in RA patients with the IFN signature. These data sug-gest that the type I IFN signature might be used to predict beforehand which patients will benefit most from rituximab treatment. Still, the predic-tive value of the type I IFN signature in itself was not sufficient to be used as a solitary biomarker. Therefore, the type I IFN signature should be as-sessed in future studies in combination with other biomarkers for its validity to guide rituximab treatment decisions.

In chapters 8 to 10 we studied the current treatment schedule of rituximab, which consists of a course of 2 times 1,000 milligram of rituximab. In B-NHL patients rituximab is less efficacious in patients with a large tumor mass and rituximab levels are lower after therapy 36-38.

In chapter 8 we analyzed whether we could find a similar relation in RA. We found that rituximab

levels were not related to the extent of B cell depletion after treatment, or to clinical response. We also found that clinical response was not significantly lower in patients who formed anti-rituximab antibodies. These data indicate that the current rituximab treatment regimen results in drug levels that are in the therapeutic range in both responders and non-responders to treat-ment, even when patients form anti-rituximab antibodies.

In chapters 9 and 10 we studied the effects of repeated treatment with rituximab in initial responders and non-responders to treat-ment. In our cohort, initial non-responders did not respond to re-treatment, while clinical responders had an improved response to a second treatment course. Our findings have recently been confirmed in a large, randomized clinical trial

39. Other studies, however, have suggested that

seropositive patients who fail to respond to a first course of rituximab may still respond to a second course 40. In light of the availability of other

therapeutic options, for individual patients who are rituximab non-responders other treatment options should be considered.

Chapter 11 describes the results of a phase Ib trial with atacicept in RA patients. Atacicept is a recombinant fusion protein that binds and neutralizes the activity of the cytokines B lymphocyte stimulator (BLyS), A proliferation-inducing ligand (APRIL) and the heterodimer of these 2 cytokines 41;42. BLyS and APRIL enhance

B cell survival, proliferation, antigen presenta-tion, and class-switch recombination at various stages of B cell development 43-44. We found that

atacicept is well tolerated with an in vivo biologic effect consistent with its mechanism of action. Treatment resulted in a decrease in serum RF and ACPA levels in parallel with total serum immuno-globulin levels. Future work will need to address whether this biological effect might translate into clinical efficacy.

MECHANISM OF ACTION OF RITUxIMAB AND BIOMARKERS OF RESPONSE. In chapter 4 to 7 we studied the effects of rituximab on synovial tissue and peripheral blood in relationship to clinical response. We identified biomarkers of response and immune mechanisms that remain active in patients who do not respond to therapy.

Rituximab induces a rapid, near com-plete depletion of CD20 positive B cells in periph-eral blood 12. In chapter 4 we analysed to which

extent rituximab depletes B cells in the synovial tissue of RA patients. We found that rituximab induces a significant decrease in synovial B cells within 4 weeks after administration, but that B cells persist in a proportion of patients. This variable depletion of synovial B cells has been confirmed by other groups 13;14. The percentage of

patients in whom synovial B cells persist differed between these studies. As discussed in chapter 5 these differences may depend on the time point at which the second synovial biopsy was taken (i.e. at 4, 12 or 16 weeks after treatment) and the choice of the specific antibodies used to detect B cells by immunohistochemistry. Similar to synovi-al tissue, it was found that B cells, especisynovi-ally with a memory phenotype, persisted in bone marrow of RA patients 15,16. Furthermore, B cells were

found to persist in lymph nodes of patients with B cell non-Hogdkin lymphoma (B-NHL) 17. In

our and other studies the persistence of synovial B cells did not predict clinical non-response to rituximab.

Conceivably, the clinical response to rituximab could be improved by achieving more complete depletion of B lineage cells in the tis-sues. This might be achieved by repeated admin-istration of rituximab in patients with incomplete response, an approach we investigated in chapter 9 (see below). Alternatively, B cell depleting agents might be developed with increased effi-cacy. A number of these drugs are currently under development. Finally, rituximab could perhaps be combined with therapeutics that enhance B cell

depletion. Up to date, no factors have been identi-fied that play a role in protecting B cells from rituximab in synovial tissue. Roles have been suggested for Fc receptor polymorphisms, expres-sion of the complement-inhibitory factors CD55 and CD59 and the B cell survival factors BlyS and CXL12, but definite prove for a protective effect has yet to be given 18-25.

In chapter 6 we analyzed the indirect effects of rituximab on synovial inflammatory cell populations and the differences in these popula-tions between responders and non-responders to treatment. We found that rituximab induces a decrease in synovial T cells and disrupts the pres-ence of SLN and follicular dendritic cells in both responders and non-responders. Furthermore, we found that rituximab induces a decrease in syno-vial macrophages, especially in the intimal lining layer. Finally, we found an indirect decrease (be-tween 4 and 16 weeks after treatment) in synovial plasma cells in clinical responders. The decrease in plasma cells was correlated with the decrease in ACPA levels in peripheral blood. Of interest, we recently found that the serum levels of free light chains, plasma cell products with a half life of a number of hours, also decrease in clinical responders to rituximab 26. Free light chain levels

are not affected by infliximab, indicating a specific indirect effect of rituximab, through depletion of CD20 positive precursors, on short lived plasma cells that produce free light chains.

In contrast to observations in clinical responders, we found that synovial plasma cells and serum free light chains persist in non-responders to rituximab treatment. Persistence of synovial B lineage cells in non-responders to rituximab was also observed in other studies 13;14. These data

sug-gest that patients may fail to respond to rituximab due to persistence of pathogenic B cells. We did not find baseline differences in the number of synovial B or plasma cells, the levels of RF, ACPA or free light chains in responders versus non-re-sponders. Of note, all patients in our study were selected on the basis of being positive for RF

Chapter 12

(5)

PAGE. 84PAGE. 85

CONCLUDING REMARKS AND FUTURE DIRECTIONS. In this thesis we investigated B cells and B cell-directed therapy in RA. We aimed to find biomarkers predictive of the clinical response to TNF block-ade and rituximab, identify immune mechanisms that remain active in patients not responding to therapy, and investigate the effect of systematic retreatment in initial responders versus non-responders. Finally, we conducted an exploratory study on the effects of atacicept in RA.

The results presented here support the notion that RA is a heterogeneous disease with a variable response to a given therapy and indicate that specific biomarkers may predict the response to therapy. The persistence of B lineage cells in the synovium of autoantibody positive RA patients who do not exhibit a robust response to the first cycle with rituximab suggests that novel ap-proaches may be needed to interfere with these cells in initial non-responders.

Chapter 12

B Cells and B Cell directed therapies in Rheumatiod Arthritis

() Manzo A, Paoletti S, Carulli M, Blades MC, Barone F, Yanni G et al. Systematic microanatomical analysis of CXCL13 and CCL21 in situ production and progressive lymphoid organization in rheuma-toid synovitis.

EUR J IMMUNOL 005; 5(5):47-59. () Weyand CM, Goronzy JJ. Ecto-pic germinal center formation in rheumatoid synovitis.

ANN N Y ACAD SCI 00;987:40–9. () Randen I, Mellbye OJ, Forre O, Natvig JB. The identification of germinal centres and follicular dendritic cell networks in rheuma-toid synovial tissue.

SCAND J IMMUNOL 995; 4(5):48-48. (4) Klimiuk PA, Goronzy JJ, Bjor nJ, Beckenbaugh RD, Weyand CM. Tis-sue cytokine patterns distinguish variants of rheumatoid synovitis.

AM J PATHOL 997; 5(5):-9. (5) Klimiuk PA, Sierakowski S, Latosiewicz R, Cylwik B, Skowron-ski J, Chwiecko J. Serum matrix metalloproteinases and tissue inhibitors of metalloproteinases in different histological variants of rheumatoid synovitis.

RHEUMATOLOGY (OxFORD) 00; 4():78-87. () Van de Sande MGH, Thurlings RM, Boumans MJH, Tak PP. The Relationship Between Synovial Lymphocyte Aggregates and

Diag-nosis and Outcome After Follow up in Early Arthritis Patients.

SUBMITTED.

(7) Cantaert T, Kolln J, Timmer T, van der Pouw Kraan TC, Vandoor-en B, Thurlings RM et al. B lympho-cyte autoimmunity in rheumatoid synovitis is independent of ectopic lymphoid neogenesis.

J IMMUNOL 008; 8():785-794.

(8) Schröder AE, Greiner A, Seyfert C, Berek C. Differentiation of B cells in the nonlymphoid tissue of the synovial membrane of patients with rheumatoid arthritis.

PROC NATL ACAD SCI USA 99; 9():-5. (9) Humby F, Bombardieri M, Man-zo A, Kelly S, Blades MC, Kirkham B et al. Ectopic lymphoid structures support ongoing production of class-switched autoantibodies in rheumatoid synovium.

PLOS MED 009; ():E.

(0) Takemura S, Klimiuk PA, Braun A, Goronzy JJ, Weyand CM. T cell activation in rheumatoid synovium is B cell dependent.

J IMMUNOL 00; 7(8):470-478. () Potter C, Hyrich KL, Tracey A, Lunt M, Plant D, Symmons DP, et al. Association of rheumatoid factor and anti-cyclic citrullinated peptide positivity, but not car-riage of shared epitope or PTPN22 susceptibility variants, with anti-tumour necrosis factor response in rheumatoid arthritis.

ANN RHEUM DIS 009 ; 8():9-74.

() Wijbrandts CA, Dijkgraaf MG,

Kraan MC, Vinkenoog M, Smeets TJ, Dinant H et al. The clinical response to infliximab in rheuma-toid arthritis is in part dependent on pretreatment tumour necrosis factor alpha expression in the synovium.

ANN RHEUM DIS 008; 7(8):9-44. () van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Rustenburg F, Baggen JM, Verweij CL et al. Responsiveness to anti-tu-mour necrosis factor alpha therapy is related to pre-treatment tissue inflammation levels in rheumatoid arthritis patients.

ANN RHEUM DIS 008; 7(4):5-5. (4) Ulfgren AK, Andersson U, Eng-strom M, Klareskog L, Maini RN, Taylor PC. Systemic anti-tumor necrosis factor α therapy in rheu-matoid arthritis down-regulates synovial tumor necrosis factor α synthesis.

ARTHRITIS RHEUM 000;4:9–.

(5) Taylor PC, Peters AM, Paleolog E, Chapman PT, Elliott MJ, McClo-skey R, et al. Reduction of chemo-kine levels and leukocyte traffic to joints by tumor necrosis factor _ blockade in patients with rheuma-toid arthritis.

ARTHRITIS RHEUM 000;4:8–47.

() Tak PP, Taylor PC, Breedveld FC, Smeets TJ, Daha MR, Kluin PM, et al. Decrease in cellular-ity and expression of adhesion molecules by anti–tumor necrosis factor _ monoclonal antibody treatment in patients with rheuma-toid arthritis.

(6)

PAGE. 8 PAGE. 87 – Chapter 12

B Cells and B Cell directed therapies in Rheumatiod Arthritis

with alemtuzumab may reverse acquired resistance to rituximab in non-Hodgkin lymphoma.

LEUK LYMPHOMA 007; 48:44-4. (4) Terui Y, Sakurai T, Mishima Y, Mishima Y, Sugimura N, Sasaoka C et al. Blockade of bulky lymphoma-associated CD55 expression by RNA interference overcomes re-sistance to complement-dependent cytotoxicity with rituximab.

CANCER SCI 00;97:7-79.

(5) Goodyear CS, Boyle DL, Silver-man GJ. Secretion of BAFF by fibroblast-like synoviocytes from rheumatoid arthritis biopsies atten-uates B cell depletion by rituximab [abstract].

ARTHRITIS RHEUM 005;5 (SUPPL 9):S90. () Radbruch A, Muehlinghaus G, Luger EO, Inamine A, Smith KG, Dörner T et al. Competence and competition: the challenge of becoming a long-lived plasma cell.

NAT REV IMMUNOL 00;(0):74-50.

(7) Fabris M, Quartuccio L, Saracco M, Pellerito R, Atzeni F, Sarzi-Puttini P, et al. BLyS Promoter Polymorphism and Response to Rituximab in Rheumatoid Arthritis (RA) Patients Positive or Negative for the Rheumatoid Factor.

ARTHRITIS RHEUM 009; 0 (0): S7. (8) Berinstein NL, Grillo-Lopez AJ, White CA, ce-Bruckler I, Maloney D, Czuczman M et al. Association of serum Rituximab (IDEC-C2B8) concentration and anti-tumor response in the treatment of recur-rent low-grade or follicular

non-Hodgkin’s lymphoma.

ANN ONCOL 998; 9(9):995-00.

(9) Igarashi T, Kobayashi Y, Ogura M, Kinoshita T, Ohtsu T, Sasaki Y et al. Factors affecting toxicity, response and progression-free survival in relapsed patients with indolent B-cell lymphoma and mantle cell lymphoma treated with rituximab: a Japanese phase II study.

ANN ONCOL 00; ():98-94.

(40) Tobinai K, Igarashi T, Itoh K, Kobayashi Y, Taniwaki M, Ogura M et al. Japanese multicenter phase II and pharmacokinetic study of rituximab in relapsed or refrac-tory patients with aggressive B-cell lymphoma.

ANN ONCOL 004; 5(5):8-80. (4) Gerlag DM, Haringman JJ, Smeets TJ, Zwinderman AH, Kraan MC, Laud PJ et al. Effects of oral prednisolone on biomarkers in synovial tissue and clinical im-provement in rheumatoid arthritis.

ARTHRITIS RHEUM 004; 50():78-79. (4) Haringman JJ, Gerlag DM, Zwinderman AH, Smeets TJ, Kraan MC, Baeten D et al. Synovial tissue macrophages: a sensitive biomark-er for response to treatment in patients with rheumatoid arthritis.

ANN RHEUM DIS 005; 4():84-88. (4) Wijbrandts CA, Vergunst CE, Haringman JJ, Gerlag DM, Smeets TJ, Tak PP. Absence of changes in the number of synovial sublin-ing macrophages after ineffective treatment for rheumatoid arthritis:

Implications for use of synovial sublining macrophages as a bio-marker.

ARTHRITIS RHEUM 007;5():89-7. (44) Bresnihan B, Pontifex E, Thurl-ings RM, Vinkenoog M, El-Gabala-wy H, Fearon U, et al. Synovial tis-sue sublining CD68 expression is a biomarker of therapeutic response in rheumatoid arthritis clinical tri-als: consistency across centers.

J RHEUMATOL. 009 AUG;(8):800-. (45) Cambridge G, Leandro MJ, Edwards JC, Ehrenstein MR, Salden M, Bodman-Smith M, et al. Serologic changes following B lymphocyte depletion therapy for rheumatoid arthritis.

ARTHRITIS RHEUM 00;48:4–54.

(4) Kormelink TG, Tekstra J, Thurl-ings RM, Boumans MH, Vos K, Tak PP, et al. Decrease in immunoglob-ulin free light chains in patients with rheumatoid arthritis upon rituximab (anti-CD20) treatment correlates with decrease in disease activity.

ANN RHEUM DIS. 00 AUG . [EPUB AHEAD OF PRINT]

(47) Crow MK. Type I interferon in systemic lupus erythematosus.

CURR TOP MICROBIOL IMMUNOL 007; :59-8.

(48) van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Voskuyl AE, Rustenburg F, Baggen JM et al. Rheumatoid arthritis subtypes identified by genomic profiling of peripheral blood cells: assignment of a type I interferon

ARTHRITIS RHEUM 99;9:077–8. (7) Tak PP, Cohen S, Emery P, Saadeh CK, De Vita S, Donohue JP, et al. Baseline Autoantibody Status (RF, Anti-CCP) and Clini-cal Response Following the First Treatment Course With Rituximab.

ARTHRITIS RHEUM 997 Sx

(8) Gong Q, Ou Q, Ye S, Lee WP, Cornelius J, Diehl L et al. Impor-tance of cellular microenvironment and circulatory dynamics in B cell immunotherapy.

J IMMUNOL 005; 74():87-8.

(9) Hamaguchi Y, Uchida J, Cain DW, Venturi GM, Poe JC, Haas KM et al. The peritoneal cavity provides a protective niche for B1 and conventional B lymphocytes during anti-CD20 immunotherapy in mice.

J IMMUNOL 005; 74(7):489-499. (0) Edwards JC, Szczepanski L, Szechinski J, Filipowicz-Sosnowska A, Emery P, Close DR et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheuma-toid arthritis.

N ENGL J MED 004; 50(5):57-58. () Kuijpers TW, Bende RJ, Baars PA, Grummels A, Derks IA, Dol-man KM, et al. CD20 deficiency in humans results in impaired T cell-independent antibody responses.

J CLIN INVEST 00; 0():4-. () Mease PJ. B cell-targeted therapy in autoimmune disease: rationale, mechanisms, and clinical application.

J RHEUMATOL 008; 5(7):45-55.

() Glennie MJ, French RR, Cragg MS, Taylor RP. Mechanisms of killing by anti-CD20 monoclonal antibodies.

MOL IMMUNOL 007; 44():8-87. (4) Kavanaugh A, Rosengren S, Lee SJ, Hammaker D, Firestein GS, Kalunian K et al. Assessment of rituximab’s immunomodulatory synovial effects (ARISE trial). 1: clinical and synovial biomarker results.

ANN RHEUM DIS 008; 7():40-408.

(5) Teng YK, Levarht EW, Hashemi M, Bajema IM, Toes RE, Huizinga TW et al. Immunohistochemical analysis as a means to predict responsiveness to rituximab treat-ment.

ARTHRITIS RHEUM 007; 5():909-98. () Cioc AM, Vanderwerf SM, Peterson BA, Robu VG, Forster CL, Pambuccian SE. Rituximab-in-duced changes in hematolymphoid tissues found at autopsy.

AM J CLIN PATHOL 008; 0(4):04-. (7) Nakou M, Katsikas G, Sidi-ropoulos P, Bertsias G, Papadimi-traki E, Raptopoulou A et al. Ritux-imab therapy reduces activated B cells both in the peripheral blood and bone marrow of patients with rheumatoid arthritis: Depletion of memory B cells correlates with clinical response.

ARTHRITIS RES THER 009; :R.

(8) Rehnberg M, Amu S, Tarkowski A, Bokarewa MI, Brisslert M. Short- and long-term effects of anti-CD20 treatment on B cell

on-togeny in bone marrow of patients with rheumatoid arthritis.

ARTHRITIS RES THER 009; (4):R. (9) Dass S, Rawstron AC, Vital EM, Henshaw K, McGonagle D, Emery P. Highly sensitive B cell analysis predicts response to rituximab therapy in rheumatoid arthritis.

ARTHRITIS RHEUM 008; 58(0):99-999. (0) Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colom-bat P et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene.

BLOOD 00; 99():754-758.

() Galimberti S, Palumbo GA, Caracciolo F, Benedetti E, Pelosini M, Brizzi S et al. The efficacy of rituximab plus Hyper-CVAD regi-men in mantle cell lymphoma is independent of FCgammaRIIIa and FCgammaRIIa polymorphisms.

J CHEMOTHER 007; 9():5-.

() Mitrovic Z, Aurer I, Radman I, Ajdukovic R, Sertic J, Labar B. FC-gammaRIIIA and FCgammaRIIA polymorphisms are not associated with response to rituximab and CHOP in patients with diffuse large B-cell lymphoma.

HAEMATOLOGICA 007; 9(7):998-999. () Cruz RI, Hernandez-Ilizaliturri FJ, Olejniczak S, Deeb G, Knight J, Wallace P et al. CD52 over-expres-sion affects rituximab-associated complement-mediated cytotoxic-ity but not antibody-dependent cellular cytotoxicity: Preclinical evidence that targeting CD52

(7)

PAGE. 88PAGE. 89 – Chapter 12

B Cells and B Cell directed therapies in Rheumatiod Arthritis

signature in a subpopulation of patients.

ANN RHEUM DIS 007; (8):008-04. (49) Bos CL, van Baarsen LG, Tim-mer TC, Overbeek MJ, Basoski NM, Rustenburg F et al. Molecular subtypes of systemic sclerosis in association with anticentromere antibodies and digital ulcers.

GENES IMMUN 009; 0():0-8.

(50) Bilgic H, Ytterberg SR, Amin S, McNallan KT, Wilson JC, Koeuth T et al. Interleukin-6 and type I interferon-regulated genes and chemokines mark disease activity in dermatomyositis.

ARTHRITIS RHEUM 009; 0():4-44. (5) van Baarsen LG, Vosslamber S, Tijssen M, Baggen JM, van d, V, Killestein J et al. Pharmacogenom-ics of interferon-beta therapy in multiple sclerosis: baseline IFN sig-nature determines pharmacologi-cal differences between patients.

PLOS ONE 008; (4):E97.

(5) Emamian ES, Leon JM, Lessard CJ, Grandits M, Baechler EC, Gaff-ney PM et al. Peripheral blood gene expression profiling in Sjogren’s syndrome.

GENES IMMUN 009; 0(4):85-9. (5) Leandro MJ, de lT, I. Trans-lational Mini-Review Series on B Cell-Directed Therapies: The pathogenic role of B cells in autoan-tibody-associated autoimmune diseases--lessons from B cell-deple-tion therapy.

CLIN ExP IMMUNOL 009; 57():9-97.

(54) Dörner T, Kinnman N, Tak PP. Targeting B cells in immune mediated inflammatory disease: A comprehensive review of mecha-nisms of action and identification of biomarkers.

PHARMACOL THER 00; 5():44-75. (55) Cantaert T, van Baarsen LGM, Wijbrandts CA, Thurlings RM, van de Sande MG, Bos C et al. Type I interferons have no major influence on humoral autoimmunity in rheu-matoid arthritis.

RHEUMATOLOGY 00; 49():5-. (5) Vital EM, Dass S, Rawstron AC, Buch MH, Goëb V, Henshaw K, Ponchel F, Emery P. Manage-ment of nonresponse to rituximab in rheumatoid arthritis: predictors and outcome of re-treatment.

ARTHRITIS RHEUM 00; (5):7-9.

(57) Vital E, Dass S, Buch MH, Atzeni F, Ponchel F, Rawstron AC, et al. A randomized double-blind placebo controlled trial on the effects of in-creased dose rituximab in patients with initial incomplete depletion – the extended treatment with rituximab in rheumatoid arthritis (EXTRRA) trial.

ARTHRITIS RHEUM 00; S xx.

(58) Marsters SA, Yan M, Pitti RM, Haas PE, Dixit VM, Ashkenazi A. Interaction of the TNF homologues BLyS and APRIL with the TNF receptor homologues BCMA and TACI.

CURR BIOL 000; 0():785-788.

(59) Gross JA, Johnston J, Mudri S, Enselman R, Dillon SR, Madden K

et al. TACI and BCMA are receptors for a TNF homologue implicated in B-cell autoimmune disease.

NATURE 000; 404(78):995-999.

(0) Moisini I, Davidson A. BAFF: a local and systemic target in auto-immune diseases.

CLIN ExP IMMUNOL 009; 58():55-. () Batten M, Groom J, Cachero TG, Qian F, Schneider P, Tschopp J et al. BAFF mediates survival of pe-ripheral immature B lymphocytes. J ExP MED 000; 9(0):45-4.

() Ye Q, Wang L, Wells AD, Tao R, Han R, Davidson A et al. BAFF binding to T cell-expressed BAFF-R costimulates T cell proliferation and alloresponses.

Referenties

GERELATEERDE DOCUMENTEN

In this chapter, I summarize my conclusions and address my initial research questions. The youth I interviewed talked at length about the important and positive aspects of their

tional shelter effectively support individuals’ transitions from homelessness to stabilized housing and reduce relapses back to the streets and emergency shelters?” One of the

Most critical to the Seattle strikers’ sense of Americanism was the assertion that labour represented the primary force responsible for bringing about positive reforms in the United

In this report, we introduced the selections of different modules for building the system economy and affordable. The system has a 980 nm central wavelength, 0.5 nm half peak width

The main objectives of the present study were (1) to inves- tigate microstructural differences in white matter between a group of individuals with SCD and matched healthy

adults who eat at least five servings of fruits and vegetables daily by 20 per cent;. • reduce the percentage

• Naïve Whittle Index Policy (NWIP): In [7], the Whittle index has been used for AoI minimization when each packet includes complete information and thus causes a status update..

participants were reaching their pubescent teenage years, The World of Suzie Wong (1960) and the Flower Drum Song (1961), featuring Nancy Kwan, a Chinese woman in the starring role,