Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Journal of Orthopaedic Science 23 (2018) 717e721

Contents lists available at ScienceDirect

Journal of Orthopaedic Science


journal homepage: http://www.elsevier.com/locate/jos

Review Article

Emerging anti-osteoclast therapy for rheumatoid arthritis*


Sakae Tanaka 1
Department of Orthopedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Rheumatoid arthritis (RA) is an autoimmune inflammatory disorder characterized by progressive
Received 18 January 2018 destruction of affected synovial joints. Recently, it was demonstrated that osteoclasts play critical roles in
Received in revised form bone destruction in RA. Receptor activator of NF-kB ligand (RANKL), which belongs to the tumor necrosis
24 March 2018
factor superfamily, is indispensable for osteoclast differentiation and bone destruction in RA. Denosu-
Accepted 2 June 2018
Available online 30 June 2018
mab, a monoclonal antibody against human RANKL, not only increased bone mineral density, but also
efficiently suppressed the progression of bone erosion in RA patients in a randomized controlled study.
However, denosumab did not reduce the cartilage destruction or disease activity in RA, and further
Keywords:
Osteoclast
investigation is required to establish the appropriate positioning of denosumab in the treatment strategy
RANKL of RA.
Rheumatoid arthritis © 2018 The Japanese Orthopaedic Association. Published by Elsevier B.V. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction 1.1. Involvement of osteoclasts in bone destruction in RA

Progressive and massive joint destruction in rheumatoid Radiographic studies have shown that bone erosion in RA
arthritis (RA) is caused by persistent synovitis, which deteriorates begins at the early stage of disease, and gradually (and some-
the activity of daily living and the quality of life of RA patients [1]. times rapidly) exacerbates. Proliferating synovium produces an
Therefore, suppressing joint destruction is one of the most impor- elevated amount of proinflammatory cytokines such as inter-
tant issues in the treatment of RA. Disease-modifying anti-rheu- leukin (IL)-1, IL-6, and tumor necrosis factor (TNF)-a, and matrix-
matic drugs (DMARDs), such as methotrexate, reduce inflammatory degrading enzymes matrix metalloproteinases and cathepsins,
synovitis in RA, and suppress joint destruction [2]. In addition, which are involved in bone and cartilage destruction [8]. Bone
recent clinical studies have demonstrated that biological DMARDs erosion usually begins at the interface of the cartilage and the
such as cytokine inhibitors both suppress disease activity and proliferating synovium (bare area), and numerous bone-
ameliorate joint destruction in RA [3]. In accordance with an resorbing osteoclasts are present at the erosive synovium/bone
improvement in disease status, a trend of decreased incidence of interface. Histochemical and ultrastructural studies by Bromley
orthopaedic surgeries in RA patients has been reported in recent and Woolley reported the presence of acid phosphatase-positive
years by many studies from different countries including Japan multinucleated cells along the surface of mineralized sub-
[4,5]. Current RA treatment has thus been considerably improved, chondral bone and mineralized cartilage [9]. These multinucle-
but it is far from perfect because these drugs are not effective in all ated cells exhibited the morphological features of osteoclasts,
patients, and their usage is sometimes associated with serious indicating osteoclasts are implicated in bone erosion in RA. More
adverse effects such as infection [6,7]. In addition, the high medical recently, Gravallese et al. demonstrated that multinucleated cells
costs of these drugs burden both patients and society. observed in the erosive surface expressed osteoclast-specific
The present review proposes that osteoclasts are involved in the genes, such as tartrate-resistant acid phosphatase and calci-
bone destruction in RA, and that a novel treatment strategy tar- tonin receptors using in situ hybridization [10]. To further un-
geting osteoclasts is an effective and safe treatment option. derstand the mechanism of osteoclast development in RA joints,
Fujikawa et al. and Takayanagi et al. developed in vitro osteoclast
formation systems using synovial cells obtained from RA patients
* [11,12]. Multinucleated cells that differentiated from synovial
This Review Article was presented at the 32nd Annual Research Meeting of the
Japanese Orthopaedic Association, Okinawa, Oct. 27, 2017. macrophages exhibited bone-resorbing activity when cultured
E-mail address: TANAKAS-ORT@h.u-tokyo.ac.jp. on bone slices. These results suggest that RA synovial tissues
1
Fax: þ81 3 5800 8858.

https://doi.org/10.1016/j.jos.2018.06.001
0949-2658/© 2018 The Japanese Orthopaedic Association. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.
org/licenses/by-nc-nd/4.0/).
718 S. Tanaka / Journal of Orthopaedic Science 23 (2018) 717e721

provide a favorable microenvironment for monocyte- progression of joint destruction in RA patients. A recent cohort
macrophage lineage cells to differentiate into osteoclasts. study demonstrated that the RANKL:OPG ratio independently
The importance of osteoclasts in RA bone destruction was predicted annual radiological damage over 11 years [22]. These
further evidenced by studies of various arthritis model animals. results strongly suggest that RANKL-induced osteoclast forma-
In mice generated by mating c-fos-deficient osteopetrosis mice, tion critically regulates bone destruction in RA, and that anti-
in which osteoclastogenesis is impaired, with human TNF-a- RANKL therapy may be useful for the prevention of bone
transgenic mice that spontaneously develop RA-like arthritis, destruction.
bone destruction does not occur despite synovial inflammation
and cartilage destruction [13]. In addition, we reported a case of 2. Clinical benefits of denosumab for osteoporosis and RA
RA associated with osteopetrosis [14], an inherited disorder
characterized by increased bone mass caused by reduced bone Denosumab is a fully human monoclonal anti-RANKL antibody
resorption. The most frequent form of osteopetrosis has auto- that specifically inhibits the interaction between RANKL and RANK,
somal dominant (ADO) inheritance (incidence 5:100,000), which and strongly suppresses osteoclast differentiation and bone
€ nberg disease or ADO type II. ADO type
is also called Albers-Scho resorption. In the pivotal Fracture Reduction Evaluation of Deno-
II results from mutations in the CLCN7 gene encoding type 7 sumab in Osteoporosis Every 6 Months (FREEDOM) study, deno-
chloride channels, and osteoclast activity is markedly reduced. sumab significantly reduced osteoporotic fractures in
We reported a very rare case of RA associated with ADO type II. postmenopausal women with osteoporosis. Denosumab signifi-
Although the patient exhibited severe inflammation and rapid cantly increased BMD and reduced the risks of vertebral, hip, and
progression of cartilage destruction, bone destruction was only non-vertebral fractures [23]. There was no increase in the risk of
developed slowly because of reduced osteoclast activity [14]. cancer, infection, cardiovascular disease, delayed fracture healing,
These results indicate the essential role of osteoclasts in bone hypocalcemia, or osteonecrosis of the jaw. In a phase III clinical trial
destruction in RA. conducted to evaluate the effect of denosumab on Japanese oste-
oporosis patients, denosumab significantly reduced the risk of new
1.2. Regulation of osteoclast development by the RANKLeRANK or worsening vertebral fracture by 65.7% at 24 months. No apparent
pathway difference in adverse events was found between denosumab and
placebo groups [24].
Receptor activator of nuclear factor kappa B ligand (RANKL) is Recently, it was demonstrated that denosumab was also effec-
a transmembrane protein belonging to the TNF superfamily tive in ameliorating bone destruction in RA. Cohen et al. investi-
[15,16]. RANKL binds to its specific receptor RANK, a member of gated joint destruction in RA patients who received placebo,
the TNF receptor superfamily, expressed in monocyte- denosumab 60 mg, or denosumab 180 mg by subcutaneous injec-
macrophage lineage osteoclast precursor cells, mature osteo- tion Q6M for 12 months. The progression in erosion score analyzed
clasts, and dendritic cells. Osteoprotegerin (OPG) is a soluble by MRI at 6 months was lower in the 60 mg denosumab group and
decoy RANKL receptor that suppresses RANKL function by significantly lower in the 180 mg denosumab group compared with
competitively inhibiting RANKL-RANK binding [15,16]. Although the placebo group. The modified Sharp erosion scores in both
RANKL was originally identified as a dendritic cell survival factor denosumab groups were significantly lower than those in the
produced by activated T cells, subsequent in vitro and in vivo placebo group at 12 months, while the progression of joint-space
studies have shown that RANKL is indispensable for the differ- narrowing or RA disease activity was unchanged [25].
entiation and activation of osteoclasts. Targeted disruption of the A phase II clinical trial was conducted to analyze the effect of
Rankl or Rank gene induced osteopetrosis in mice because of the denosumab in Japanese RA patients with a disease duration of 6
lack of osteoclasts, while knock-out of the Opg gene induced months to less than 5 years [26]. Patients were randomly assigned
severe osteoporosis via a marked increase in osteoclasts, indi- to receive one of four treatments: placebo, denosumab 60 mg Q6M,
cating the essential role of the RANKL-RANK pathway in the Q3M or every Q2M. While denosumab significantly inhibited the
differentiation of osteoclasts from precursor cells [15]. Numerous progression of bone erosion at 12 months at all doses compared
studies have shown that the RANKL-RANK system is implicated with placebo, it had no protective effect on joint-space narrowing,
in physiological bone resorption, and also plays an important role which was consistent with the results reported by Cohen et al.
in pathological bone destruction [16]. (Fig. 2). There was no apparent difference in the safety profiles of
The important role of the RANKL/RANK/OPG system in the denosumab and placebo, including infection. All denosumab
development of bone destruction in RA has been recently groups significantly increased lumbar spine and total hip BMD
established [17] (Fig. 1). Several groups simultaneously reported compared with the placebo group at 6 and 12 months. These results
that RANKL is highly expressed in the synovial tissues of RA provide strong evidence to suggest denosumab prevents the pro-
patients [17e19]. The cells expressing RANKL are considered to gression of bone erosion in the early stage of RA, but has little or no
be activated T lymphocytes and/or synovial fibroblastic cells. effect on cartilage deterioration or disease activity. A subsequent
Kong et al. reported that when murine bone marrow precursors phase III clinical trial showed that denosumab 60 mg Q6M and Q3M
were cocultured with activated CD4þ T cells fixed with para- significantly suppressed bone erosion at 12 months in Japanese RA
formaldehyde, osteoclasts were differentiated in vitro [20]. We patients [27]. Based on these results, denosumab was approved for
found that in a coculture system containing synovial fibroblasts “inhibition of the progression of bone erosion associated with RA”
and peripheral monocytes, 1a,25-dihydroxyvitamin D3 treatment in Japan.
induced RANKL expression in synovial fibroblasts and induced Denosumab has additional benefits in RA patients by preventing
osteoclast differentiation from monocytes [19]. Kong et al. also osteoporosis and osteoporotic fractures. RA patients are at
reported that blocking RANKL by OPG treatment prevented joint increased risk of osteoporotic fractures. Although the proportion of
destruction but not inflammation in adjuvant arthritis rats. In RA patients who achieve remission has greatly increased, recent
addition, Pettit et al. reported that RANKL knockout mice were studies reported the number of fractures in RA patients was not
protected from bone erosion in a serum transfer model of reduced [28,29]. This discrepancy is at least partly accounted for by
arthritis [21]. Several clinical studies have shown that the RAN- the multifactorial nature of osteoporosis in RA patients, with
KL:OPG ratio in serum or synovial fluid may predict the contributory factors other than inflammation, such as immobility,
S. Tanaka / Journal of Orthopaedic Science 23 (2018) 717e721 719

Fig. 1. Regulation of osteoclast differentiation and bone erosion in rheumatoid arthritis by RANKL. M-CSF: macrophage colony-stimulating factor.

Fig. 2. Denosumab significantly suppresses bone erosion score (a) but has no effect on joint-space narrowing score (b) Modified from a Figure in the article by Takeuchi et al. [26].
CI: confidence interval.
720 S. Tanaka / Journal of Orthopaedic Science 23 (2018) 717e721

Fig. 3. Schematic representation of the possible mechanisms of joint destruction in RA. Modified from a Figure in the article by Tanaka et al. [30].

steroid usage, and aging. Clinical studies to analyze the effect of van Eijk-Hustings Y, Emery P, Finckh A, Gabay C, Gomez-Reino J, Gossec L,
Gottenberg JE, Hazes JMW, Huizinga T, Jani M, Karateev D, Kouloumas M,
denosumab on RA patients also demonstrated an increase in BMD
Kvien T, Li Z, Mariette X, McInnes I, Mysler E, Nash P, Pavelka K, Poor G,
[25,26]. Although it remains unknown whether denosumab treat- Richez C, van Riel P, Rubbert-Roth A, Saag K, da Silva J, Stamm T, Takeuchi T,
ment can reduce fragility fractures in RA patients, anti-osteoporosis Westhovens R, de Wit M, van der Heijde D. EULAR recommendations for the
treatment is absolutely required for these patients, and denosumab management of rheumatoid arthritis with synthetic and biological disease-
modifying antirheumatic drugs: 2016 update. Ann Rheum Dis 2017
may play a useful role. Fig. 3 summarizes the possible role of Jun;76(6):960e77.
denosumab in the treatment of RA. [4] Momohara S, Tanaka S, Nakamura H, Mibe J, Iwamoto T, Ikari K, Nishino J,
In conclusion, recent studies demonstrated that denosumab is Kadono Y, Yasui T, Takahashi K, Takenouchi K, Hashizume K, Nakahara R,
Kubota A, Nakamura T, Nishida K, Suguro T. Recent trends in orthopedic
effective in ameliorating bone destruction in RA. However, it surgery performed in Japan for rheumatoid arthritis. Mod Rheumatol 2011
should be noted that denosumab has no effect on joint inflam- Aug;21(4):337e42.
mation or cartilage destruction in RA, and should therefore be [5] Matsumoto T, Nishino J, Izawa N, Naito M, Hirose J, Tanaka S, Yasui T, Saisho K,
Tohma S. Trends in treatment, outcomes, and incidence of orthopedic surgery
used in combination with other therapeutics. The optimal posi- in patients with rheumatoid arthritis: an observational cohort study using the
tioning of denosumab in the treatment strategy of RA remains Japanese national database of rheumatic diseases. J Rheumatol 2017
elusive, and many clinical questions remain unanswered. Further Nov;44(11):1575e82.
[6] Ramiro S, Sepriano A, Chatzidionysiou K, Nam JL, Smolen JS, van der Heijde D,
investigation is necessary to answer these questions and to Dougados M, van Vollenhoven R, Bijlsma JW, Burmester GR, Scholte-
establish the appropriate position of denosumab in the treatment Voshaar M, Falzon L, Landewe RBM. Safety of synthetic and biological
strategy of RA. DMARDs: a systematic literature review informing the 2016 update of the
EULAR recommendations for management of rheumatoid arthritis. Ann
Rheum Dis 2017 Jun;76(6):1101e36.
Conflict of interest [7] Komano Y, Tanaka M, Nanki T, Koike R, Sakai R, Kameda H, Nakajima A,
Saito K, Takeno M, Atsumi T, Tohma S, Ito S, Tamura N, Fujii T, Sawada T,
Ida H, Hashiramoto A, Koike T, Ishigatsubo Y, Eguchi K, Tanaka Y,
None. Takeuchi T, Miyasaka N, Harigai M, Group RS. Incidence and risk factors for
serious infection in patients with rheumatoid arthritis treated with tumor
Acknowledgements necrosis factor inhibitors: a report from the Registry of Japanese Rheuma-
toid Arthritis Patients for Longterm Safety. J Rheumatol 2011 Jul;38(7):
1258e64.
The work of S. Tanaka is supported by The Japan Agency for [8] Firestein GS. Evolving concepts of rheumatoid arthritis. Nature 2003 May
Medical Research and Development (AMED) (924887), Japan So- 15;423(6937):356e61.
[9] Bromley M, Woolley DE. Chondroclasts and osteoclasts at subchondral sites of
ciety for the Promotion of Science (JSPS)/Grant-in-Aid for Scientific erosion in the rheumatoid joint. Arthritis Rheum 1984 Sep;27(9):968e75.
Research (A) (406501), and Japan Society for the Promotion of [10] Gravallese EM, Harada Y, Wang JT, Gorn AH, Thornhill TS, Goldring SR.
Science (JSPS)/Grant-in-Aid for Exploratory Research (934289). Identification of cell types responsible for bone resorption in rheumatoid
arthritis and juvenile rheumatoid arthritis. Am J Pathol 1998 Apr;152(4):
943e51.
References [11] Fujikawa Y, Sabokbar A, Neale S, Athanasou NA. Human osteoclast formation
and bone resorption by monocytes and synovial macrophages in rheumatoid
[1] Schett G. Autoimmunity as a trigger for structural bone damage in rheumatoid arthritis. Ann Rheum Dis 1996 Nov;55(11):816e22.
arthritis. Mod Rheumatol 2017 Mar;27(2):193e7. [12] Takayanagi H, Oda H, Yamamoto S, Kawaguchi H, Tanaka S, Nishikawa T,
[2] Wassenberg S, Rau R. Radiographic healing with sustained clinical remission Koshihara Y. A new mechanism of bone destruction in rheumatoid arthritis:
in a patient with rheumatoid arthritis receiving methotrexate monotherapy. synovial fibroblasts induce osteoclastogenesis. Biochem Biophys Res Commun
Arthritis Rheum 2002 Oct;46(10):2804e7. 1997 Nov 17;240(2):279e86.
[3] Smolen JS, Landewe R, Bijlsma J, Burmester G, Chatzidionysiou K, Dougados M, [13] Redlich K, Hayer S, Ricci R, David JP, Tohidast-Akrad M, Kollias G, Steiner G,
Nam J, Ramiro S, Voshaar M, van Vollenhoven R, Aletaha D, Aringer M, Smolen JS, Wagner EF, Schett G. Osteoclasts are essential for TNF-alpha-
Boers M, Buckley CD, Buttgereit F, Bykerk V, Cardiel M, Combe B, Cutolo M, mediated joint destruction. J Clin Invest 2002 Nov;110(10):1419e27.
S. Tanaka / Journal of Orthopaedic Science 23 (2018) 717e721 721

[14] Kadono Y, Tanaka S, Nishino J, Nishimura K, Nakamura I, Miyazaki T, [24] Nakamura T, Matsumoto T, Sugimoto T, Hosoi T, Miki T, Gorai I, Yoshikawa H,
Takayanagi H, Nakamura K. Rheumatoid arthritis associated with osteopet- Tanaka Y, Tanaka S, Sone T, Nakano T, Ito M, Matsui S, Yoneda T, Takami H,
rosis. Mod Rheumatol 2009;19(6):687e90. Watanabe K, Osakabe T, Shiraki M, Fukunaga M. Clinical Trials Express:
[15] Theill LE, Boyle WJ, Penninger JM. RANK-L and RANK: T cells, bone loss, and fracture risk reduction with denosumab in Japanese postmenopausal women
mammalian evolution. Annu Rev Immunol 2002;20:795e823. and men with osteoporosis: denosumab fracture intervention randomized
[16] Tanaka S, Nakamura K, Takahasi N, Suda T. Role of RANKL in physiological and placebo controlled trial (DIRECT). J Clin Endocrinol Metab 2014 Jul;99(7):
pathological bone resorption and therapeutics targeting the RANKL-RANK 2599e607.
signaling system. Immunol Rev 2005 Dec;208:30e49. [25] Cohen SB, Dore RK, Lane NE, Ory PA, Peterfy CG, Sharp JT, van der Heijde D,
[17] Gravallese EM, Manning C, Tsay A, Naito A, Pan C, Amento E, Goldring SR. Zhou L, Tsuji W, Newmark R, Denosumab Rheumatoid Arthritis Study G.
Synovial tissue in rheumatoid arthritis is a source of osteoclast differentiation Denosumab treatment effects on structural damage, bone mineral density,
factor. Arthritis Rheum 2000 Feb;43(2):250e8. and bone turnover in rheumatoid arthritis: a twelve-month, multicenter,
[18] Shigeyama Y, Pap T, Kunzler P, Simmen BR, Gay RE, Gay S. Expression of randomized, double-blind, placebo-controlled, phase II clinical trial. Arthritis
osteoclast differentiation factor in rheumatoid arthritis. Arthritis Rheum 2000 Rheum 2008 May;58(5):1299e309.
Nov;43(11):2523e30. [26] Takeuchi T, Tanaka Y, Ishiguro N, Yamanaka H, Yoneda T, Ohira T, Okubo N,
[19] Takayanagi H, Iizuka H, Juji T, Nakagawa T, Yamamoto A, Miyazaki T, Genant HK, van der Heijde D. Effect of denosumab on Japanese patients with
Koshihara Y, Oda H, Nakamura K, Tanaka S. Involvement of receptor activator rheumatoid arthritis: a dose-response study of AMG 162 (Denosumab) in
of nuclear factor kappaB ligand/osteoclast differentiation factor in osteoclas- patients with RheumatoId arthritis on methotrexate to Validate inhibitory
togenesis from synoviocytes in rheumatoid arthritis. Arthritis Rheum 2000 effect on bone Erosion (DRIVE)-a 12-month, multicentre, randomised, double-
Feb;43(2):259e69. blind, placebo-controlled, phase II clinical trial. Ann Rheum Dis 2016
[20] Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, Capparelli C, Li J, Jun;75(6):983e90.
Elliott R, McCabe S, Wong T, Campagnuolo G, Moran E, Bogoch ER, Van G, [27] Takeuchi T, Tanaka Y, Soen S, Yamanaka H, Yoneda T, Tanaka S, Nitta T,
Nguyen LT, Ohashi PS, Lacey DL, Fish E, Boyle WJ, Penninger JM. Activated T Okubo N, Genant HK, Van der Heijde D. Effects of denosumab, a subcutaneous
cells regulate bone loss and joint destruction in adjuvant arthritis through RANKL inhibitor, on the progression of structural damage in Japanese patients
osteoprotegerin ligand. Nature 1999 Nov 18;402(6759):304e9. with rheumatoid arthritis treated with cs DMARDs: results from the 12-
[21] Pettit AR, Ji H, von Stechow D, Muller R, Goldring SR, Choi Y, Benoist C, month double blind phase 3, DESIRABLE study. Ann Rheum Dis
Gravallese EM. TRANCE/RANKL knockout mice are protected from bone 2017;76(supple 2):841.
erosion in a serum transfer model of arthritis. Am J Pathol 2001 Nov;159(5): [28] Ochi K, Inoue E, Furuya T, Ikari K, Toyama Y, Taniguchi A, Yamanaka H,
1689e99. Momohara S. Ten-year incidences of self-reported non-vertebral fractures in
[22] van Tuyl LH, Voskuyl AE, Boers M, Geusens P, Landewe RB, Dijkmans BA, Japanese patients with rheumatoid arthritis: discrepancy between disease
Lems WF. Baseline RANKL: OPG ratio and markers of bone and cartilage activity control and the incidence of non-vertebral fracture. Osteoporos Int
degradation predict annual radiological progression over 11 years in rheu- 2015 Mar;26(3):961e8.
matoid arthritis. Ann Rheum Dis 2010 Sep;69(9):1623e8. [29] Kim SY, Schneeweiss S, Liu J, Solomon DH. Effects of disease-modifying
[23] Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, antirheumatic drugs on nonvertebral fracture risk in rheumatoid arthritis: a
Zoog HB, Austin M, Wang A, Kutilek S, Adami S, Zanchetta J, Libanati C, population-based cohort study. J Bone Miner Res 2012 Apr;27(4):789e96.
Siddhanti S, Christiansen C, Trial F. Denosumab for prevention of fractures in [30] Tanaka S, Tanaka Y, Ishiguro N, Yamanaka H, Takeuchi T. RANKL: a therapeutic
postmenopausal women with osteoporosis. N Engl J Med 2009 Aug 20;361(8): target for bone destruction in rheumatoid arthritis. Mod Rheumatol 2018
756e65. Jan;28(1):9e16.

You might also like