You are on page 1of 8

Joint Bone Spine 88 (2021) 105164

Contents lists available at ScienceDirect

Joint Bone Spine


journal homepage: www.elsevier.com

Review

Antigen-specific tolerance approach for rheumatoid arthritis: Past,


present and future
Audrey Page , Floriane Fusil , François-Loïc Cosset ∗
CIRI – Centre international de recherche en infectiologie, Université de Lyon, Université Claude-Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon,
46, allée d’Italie, 69007 Lyon, France

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

Article history: Rheumatoid arthritis is a chronic systemic autoimmune disease, affecting mainly the joints. It is caused
Accepted 2 February 2021 by an adaptive immune reaction against self-antigens, leading to the over production of inflammatory
Available online 19 February 2021 cytokines and autoantibodies, mainly mediated by autoreactive CD4+ T cells and pathological B cell
clones. The treatment options currently available rely on palliative global immunosuppression and do
Keywords: not restore tolerance to self-components. Here, we review antigen-specific tolerance approaches that
Rheumatoid arthritis have been developed to inhibit or delete autoreactive clones, while maintaining a potent immune sys-
Antigen-specific tolerance
tem for rheumatoid arthritis. The first attempts relied on the oral ingestion of self-reactive peptides,
Self-peptides
Tolerogenic cells
with lukewarm results in human clinical trials. To enhance treatment efficacy, self-peptides have been
Anergy engineered and combined with immunosuppressive molecules. In addition, several routes of delivery
have been tested, in particular, nanoparticles carrying self-antigens and immunomodulatory molecules.
More recently, transfer of immune cells, such as tolerogenic dendritic cells or regulatory T cells, has been
considered to restore tolerance. Although promising results have been obtained in mouse models, the
translation to humans remains highly challenging, mainly because the disease is already well developed
when treatments start and because patient’s specific self-antigens are often unknown. Nevertheless,
these approaches hold great promises for long-term RA treatment.
© 2021 Société française de rhumatologie. Published by Elsevier Masson SAS. All rights reserved.

1. Introduction This inflammation is caused by a rupture of tolerance. Even if the


initial trigger of RA is not clear and surely multifactorial, genetic,
Rheumatoid arthritis (RA) is an autoimmune disease that affects environmental factors, smoking and buccal infections by several
ca. 0.5% to 1% of the population worldwide. Typical patient symp- pathogens have been associated to breakdown of tolerance [3,4].
toms range from morning stiffness, pain, and difficulties to walk RA patients have defective central and peripheral tolerance check-
to life-threatening complications, such as cardiac or pulmonary points, which may promote the development of auto-reactive B and
issues. The disease is characterised by an immune reaction against T-cell clones that escape the negative selection mechanisms and are
self-antigens that leads to chronic systemic inflammation. The detected in RA patients [5]. These autoreactive B cells actively pro-
synovial tissue of joints is particularly inflamed, which trigg- duce pathological autoantibodies that can be used as disease and
ers subsequent erosion of cartilage and bone driven by matrix prognosis biomarkers [6]. For RA, autoantibodies are divided into
metalloproteinases, produced locally by macrophages and syno- two main categories: rheumatoid factors (RFs), which recognise
vial fibroblasts following pro-inflammatory cytokines exposure the Fc portion of immunoglobulins, and anti-citrullinated protein
[1]. Interestingly, anti-inflammatory cytokines, such as IL-10 and antibodies (ACPAs) [7]. ACPAs recognise post-translationally modi-
TGF-␤, can counteract pro-inflammatory action, by inhibiting fied self-proteins, including vimentin, filaggrin, collagen, fibrinogen
MMP production and also by inducing native inhibitors of MMPs and tenascin-C, in which arginine residues have been converted
[2]. to citrulline [8,9]. Although citrullination of proteins is a nor-
mal process that is catalysed by the peptidyl-arginase deiminase
(PAD) enzyme, the development of autoimmunity against these
citrullinated antigens marked by ACPAs is not normal [10]. Of
∗ Corresponding author. note, activated PADs have been shown to be released in neu-
E-mail address: flcosset@ens-lyon.fr (F.-L. Cosset). trophil extracellular traps (NETs) [10]. NETs are DNA filaments

https://doi.org/10.1016/j.jbspin.2021.105164
1297-319X/© 2021 Société française de rhumatologie. Published by Elsevier Masson SAS. All rights reserved.
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

entangled in proteins that are released in the extracellular Here, we review the main approaches that have been under-
space [10]. They are produced by polymorphonuclear neutrophils taken over the past years to restore antigen-specific tolerance in
and are believed to display pro-inflammatory molecules and RA.
auto-antigens (that can be citrullinated in situ), which would
enhance epitope spreading and autoimmunity, thus worsening the
disease [10]. 2. Native self-peptides administration
Among auto-antigens associated to RA, one of the most stud-
ied is type II collagen (Collagen II). Anti-collagen II autoantibodies Oral tolerisation is probably the most studied approach for
directed either against native triple helical or citrullinated epi- restoration of antigen-specific tolerance since the first attempts
topes are present in the serum, synovial fluid and cartilage of date from the beginning of the 1990s (Fig. 1). This approach
respectively 10% and 40% of RA patients [11]. The immunisation takes advantage of the natural tolerisation processes occurring
of mice with collagen II leads to pathological manifestations that when food is ingested, which prevents allergy in physiological
mimic several aspects of RA and represents a model called collagen- context. Several barriers restrict macromolecule ingestion across
induced-arthritis (CIA) that is frequently used. Although the exact the intestinal epithelium, such as proteases and mucus. M cells,
pathogenic role of citrullinated collagen II in human RA patients which are located in the Peyer’s patches of the gut-associated lym-
is not known, this CIA model has been widely used to study RA phoid tissue (GALT), are specialised in the transport of antigens
pathogenesis and to develop new treatments. by transcytosis across the mucosal barrier. After transcytosis, anti-
Current treatments for RA rely on global immunosuppressant gens are loaded on antigen-presenting cells, such as dendritic cells
drugs to control inflammation by targeting downstream effectors (DCs), and may interact with other immune cells, such as T cells.
of autoimmune responses. Non-steroidal anti-inflammatory drugs These cells then migrate to mesenteric lymph nodes and reach the
(NSAIDs), glucocorticoids or disease-modifying anti-rheumatic systemic immune circulation before being recruited at inflamed
drugs (DMARDs), such as methotrexate, are usually indicated as areas. Depending on the dose of oral self-peptide intake, different
first-line medications [12]. New treatments have driven significant tolerance mechanisms can be induced: anergy of autoreactive T
improvements, such as anti-inflammatory drugs, anti-cytokine cells, deletion of peripheral antigen-specific T cells, or induction
therapies, monoclonal antibodies (anti-TNF␣), and T-cell or B cell of Tregs, for high or low doses of antigens respectively [14,15].
inhibitors, and, recently, inhibitors of cytokine signalling (JAK Indeed, stimulation with high doses of self-reactive peptides blocks
inhibitors) [12]. Although these therapeutic molecules alleviate T-cell proliferation and IL-2 production by immune cells or cause
patient’s symptoms, and consequently improve their quality of life, activation-induced cell death. In contrast, low peptide doses may
they do not address the causes of the disease. Consequently, nei- lead to antigen-driven suppression of autoreactive clones by induc-
ther long-term drug-free remission nor restoration of tolerance to tion of Tregs in the GALT, which then migrate to the systemic
self-antigens can be achieved, and lifelong treatments are required. immune system and to inflamed areas, such as the osteoarticular
Therefore, there is an obvious need for new RA treatments that joints [14–16]. These cells mediate their immunosuppressive func-
could specifically target the causes of the disease, thus reducing tions through secretion of anti-inflammatory cytokine, such as IL-4,
side effects. In this context, re-inducing self-antigen-specific toler- IL-10 and TGF-␤. Interestingly for autoimmune disease therapy,
ance has emerged as an attractive strategy to control both cellular low-dose self-antigen intake may also suppress immune responses
and humoral autoimmune responses. unrelated to the ingested self-peptide by bystander effects, since
Importantly, an ideal immunotherapy cure would reprogram Tregs primed in an Ag-specific manner can drive Ag-unspecific
the immune system to a state of self-tolerance and lead to disease suppressions [16].
remission by specifically impacting pathological immune reactions Several self-antigens, such as bovine or chicken collagen II,
and cells. Indeed, a potent protective immunity against pathogens HCgp39 and dnaJp1, have been investigated for restoration of oral
and cancer cells, for instance, should be permanently maintained tolerance in RA. Ingestion of collagen II peptides or whole colla-
during and after the treatment. Such approaches aim mainly at gen II proteins markedly reduced arthritis incidence and severity in
modulating T cell subpopulations, either by depleting autoreactive collagen-induced arthritis (CIA) mice [14,17,18]. While most stud-
CD4+ Th1 and Th17 cells or by inhibiting their effector functions ies have focused on prophylactic effects of oral treatment, i.e. by
through the expansion of self-antigen-specific regulatory T cells starting it before or concomitantly to animal immunisation, one
(Tregs). The ratio of Treg and Th17 cells has emerged as a new study showed a beneficial impact on disease severity in a ther-
paradigm for RA [13]. Indeed, although Treg counts are heteroge- apeutic setting, paving the way for human diseases translation
nous in RA patients, their function is impaired and, hence, they fail [18]. However, results from human clinical trials after self-peptide
to inhibit the activation of pathologic immune cells, notably T cells ingestion have been quite disappointing so far and only led to
[13]. In addition, autoreactive B cells have key pathological func- minor improvements of rheumatic scores as compared to control
tions, not only as antigen-presenting cells, but also by mediating or placebo treatment [19–24].
direct inhibition of autoreactive T cells or conversion to regulatory Further clinical trials are required to assess the efficacy of these
B cells, hence resulting in decreased amounts of autoantibodies. approaches. Nevertheless, oral treatments with self-antigen pep-
This would also reverse the ratio of pro-inflammatory cytokines, tides are globally well tolerated by patients with minor adverse
such as IL-1 and TNF-␣ over anti-inflammatory cytokines, such as events, and their efficacy might be enhanced by determining the
IL-10 and TGF-␤, and ameliorate disease manifestations. Indeed, optimal doses, frequency of injection, formulation of self-peptides,
these two latter cytokines can induce Tregs and promote tissue and by combination with current anti-rheumatic drugs.
repair. Even if oral ingestion is a very convenient way for
However, some limitations are intrinsically related to such tech- self-peptide delivery, several other routes of administra-
nologies. Indeed, even if some self-antigens commonly associated tion have been tested, such as direct gastric application of
to RA are known, the diseases causing self-antigens are often dif- self-peptides [25,26] or nasal application [27,28]. The more com-
ferent among patients and evolve by epitope spreading during the monly used route to deliver most vaccines is via intradermal
course of the disease, so the treatments need to be adapted. Nev- injections, which also showed particularly interesting results
ertheless, antigen-specific tolerance approaches hold promise for for CIA [29]. The intravenous route has also been considered for
longer-term efficacy, greater specificity and lower toxicity for RA self-peptides infusion and induced a state of self-tolerance after
treatment. immunisation [30]. One potential explanation for this lack of

2
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

Fig. 1. Mechanisms of oral tolerance in RA. Self-peptides or proteins are orally given to the patient. In the GALT (gut-associated lymphoid tissue), and more particularly in
Peyer’s patches, self-peptides are transcytosed by M cells, and loaded on antigen presenting cells (APCs), such as dendritic cells. Autoreactive T cells interact with antigen
loaded APCs and may either undergo anergy or deletion or will differentiate into regulatory T cells. These Tregs migrate to mesenteric lymph nodes, where they can reach
other immune cells and inhibit the autoreactive clones. Autoreactive regulatory cells (B and T cells) then regulate the autoimmune inflammation at the joints by inhibiting
autoantibody secretion and restoring the balance between Th1/17 and Treg/Th2 that respectively secrete pro and anti-inflammatory cytokines.

clinical efficacy might be the short half-life of self-peptides and, disease severity in a therapeutic approach [35–37]. These bene-
consequently, several additional injections might be required to ficial effects were associated to a decrease in collagen II-specific
reach a therapeutic effect. antibodies and an increase in the percentage of Tregs [35,36].
Although the induction of self-peptides in vivo to restore toler-
ance seems a promising approach, to date, this strategy cannot be
3. Gene therapy widely developed for RA, especially because the post-translational
modifications of self-peptides, such as citrullination, cannot be
One of the main hurdles with the above-mentioned approaches encoded genetically. Consequently, these peptides need to be
is that repeated injections of high doses of tolerogen molecules, exogenously produced with the correct modifications and then
such as native self-peptides, might be required. This represents infused. Before infusion, these self-peptides can also be engineered
elevated costs and inconvenience to patients, due to recurrent pro- to enhance their tolerogenic properties or to promote their delivery
cedures. To circumvent these issues, several strategies have been to the right cells and at the right place.
developed to induce in vivo expression of tolerogenic self-peptides.
Firstly, DNA vaccines encoding collagen II yielded signifi-
cant improvements in CIA mouse models after intramuscular or
intravenous injection [31,32]. Joint destruction, pro-inflammatory 4. Infusion engineered bio-compounds
cytokines and epitope spreading have been significantly decreased
following such vaccination [31]. 4.1. Self-peptide fusion molecules
Alternatively, viral vectored gene therapy with lentivirus, ade-
novirus or vaccina virus has been applied to express in vivo Several concepts have been tested to enhance the tolerogenic
autoantigen and cure RA [33–37]. For instance, the intravenous potential of infused self-peptides, either by modifying their epi-
infusion of lentiviral vectors, encoding the immuno-dominant epi- topes to render them more immunosuppressive, by combining
tope of CII in the CLIP peptide, not only prevented the development several self-peptides together or by fusing them with immunosup-
of the pathology in a prophylactic setting, but also greatly reduced pressive moieties.

3
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

Some native regions of self-proteins have been shown to impairing their clinical success, is the route of delivery. Indeed,
exhibit immunosuppressive properties, such as a portion of bovine following injection, such molecules are rapidly degraded. Nanopar-
collagen II that induces an immune response against an immuno- ticles appear as promising platforms for drug delivery and have
dominant region of the TCR [29]. However, self-peptides can also been tested for self-peptide carriage (and protection from inhi-
be engineered in order to suppress pathological immune responses bition by circulating autoantibodies), to restore tolerance in RA.
while maintaining antigen specificity. For instance, one study Several approaches based on nanoparticles have been explored for
reported a recombinant collagen II peptide harbouring three point re-induction of tolerance (Fig. 2).
mutations (in residues critical for antigen presentation and thus Firstly, antigen-presenting cells can be targeted by nanopar-
T-cell activation), which drastically suppressed immunity to colla- ticles as nanoparticles are rapidly cleared by innate immune
gen II after co-immunisation with native collagen II [38] or after phagocytes. For instance, the oral ingestion of polymer contain-
intra-articular tolerogen peptide gene transfer [34]. ing collagen II has greatly reduced the incidence and the severity
As an alternative approach, multi-epitope fusion peptides have of CIA, as well as the autoantibody titers, compared to control
been created, which expands the array of epitopes against which groups [43]. Treatment efficacy may be further enhanced by com-
tolerance is sought and counteracts epitope spreading, a process bining immune-modulators on the nanoparticle, such as cytokines,
that widens the range of closely related self or not-self peptides toll-like-receptor (TLR) ligands or rapamycin, which can promote
recognised by the immune system. Such multi-epitope molecules cell differentiation toward more tolerogenic phenotypes. Remark-
can be composed of several epitopes from the same protein, such as, ably, the co-delivery of NF-␬b inhibitors and self-peptides loaded
e.g. a recombinant peptide containing two tolerogenic epitopes of on liposomes reduced clinical signs of the disease in mice [44].
chicken collagen II [39], or of epitopes from different self-proteins Similarly, calcitriol addition in liposomes has shown encouraging
[40]. For instance, a synthetic fusion protein incorporating cit- results in a proteoglycan (PG)-induced mouse model, as it sup-
rullinated epitopes of vimentin, fillagrin, fibrinogen and collagen pressed the expansion and function of Teffs, and induced Tregs
II reduced arthritis severity to a much greater extent than the [45].
mono-epitope peptide [40]. This was mediated by decreased infil- Secondly, autoreactive B and T lymphocytes can be directly
tration of immune cells in the joints and by generation of Tregs targeted and inhibited. For inhibition of autoreactive B cell, self-
[40]. peptides can be directly bound on the nanoparticle surface along
Finally, immuno-modulators, such as tolerogen molecules or with cell-targeting and immunosuppressive ligands. For example,
activation inhibitors, e.g. immune checkpoints inhibitors, have the coupling of citrullinated self-peptides with CD22, an immune
appeared as good candidates for linkage to self-antigens. Fusion inhibitor [46], or with complement-activating lytic peptide [47]
molecules have been created to exploit the tolerogenic potential of was shown to selectively inhibit pathological B cells. For inhibition
antigen presentation either by direct targeting (with anti-DEC205 of autoreactive T cell, nanoparticles can harbour MHC complexes on
antibodies to induce its delivery to DCs) [41] or by compelling pre- the nanoparticle surface that present self-peptides in the absence
sentation on MCH II (with linkage to human ␤2 microglobulin) of co-stimulation, which may induce suppression of effector
[42]. function.
Overall, nanoparticles represent an auspicious method to inhibit
4.2. Nanoparticles autoimmune responses either by acting directly on pathologic lym-
phocytes or by presentation on APCs; yet, clinical trials are required
Although molecular engineering has greatly improved the to test their safety and efficacy at a large scale before this therapeu-
tolerogenic potential of self-peptide drugs, a major limitation tic approach become widely available.

Fig. 2. Antigen-specific tolerance restoration by nanoparticles. Two main approaches for restoring tolerance can be used with nanoparticles. Firstly, APCs can be targeted.
In this case, nanoparticles contain or harbour self-peptides (which will be phagocytosed and presented on MHC-II molecules), sometimes in combination with immuno-
modulators (which will inhibit pathologic inflammation), restoring tolerance to self-peptides. Secondly, autoreactive adaptive lymphocytes can be targeted. For autoreactive
T cell inhibition, nanoparticles can harbour MCH-self-antigen complex with checkpoints co-inhibitors, while for autoreactive B cell inhibition, self-peptides can be directly
bound to nanoparticle surface. Although nanoparticles are naturally endocytosed by phagocytes, antibodies or receptors recognising cell surface molecules can be added on
the nanoparticles to target specific cell types.

4
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

5. Infusion of engineered immune cell In the two above-mentioned clinical trials, the intradermal
and the subcutaneous routes were respectively tested, while the
5.1. Engineering dendritic cells AuToDeCRA trial assessed the safety of another route for tolDC
injection, namely by intra-articular administration [53]. TolDCs
Besides nanoparticles, cells may also directly be used to carry were differentiated from patient’s monocytes and pulsed with
self-antigens, after ex vivo loading and re-infusion. DCs repre- synovial fluids containing self-antigens, before injection in the
sent favourite targets for such purposes as they are professional inflamed knee [53]. Although a decrease of arthroscopic symp-
antigen-presenting cells that naturally migrate from peripheral toms was observed in some patients, no systemic effects could be
tissue to lymph nodes, where they interact with T cells through detected [53].
the presentation of antigen in a human leukocyte antigen (HLA) Overall, several types of self-antigens, doses of infused cells
– restricted manner. This interaction can either activate or inhibit and routes of injection have been tested in phase I clinical tri-
immune responses against the presented antigen, depending on als, demonstrating the safety of such approaches but only driving
the DC phenotype. Immature and mature endogenous DCs can- minor or no improvements in rheumatic patients (Table 1). These
not be directly re-injected after self-peptide loading, since they limited improvements of the clinical scores might result from
would actually cause immunogenicity instead of tolerance. Sev- the status of these autoimmune arthritis patients, in whom dys-
eral approaches have been explored to skew maturation of DCs regulated immune responses are already established, sometimes
toward induction of a tolerogenic state (TolDCs) ex vivo before for years. Further studies are required to really assess the clin-
their reinfusion by specific culture conditions [48]. These tolero- ical efficacy and antigen-specific effects of TolDC therapy in RA
genic effector functions ameliorate RA disease manifestations patients. Interestingly, combinations with anti-rheumatic drugs
after adoptive transfer in mouse models through reduction of may act synergistically and potentiate treatments with TolDCs
Th17 cells, suppression of autoreactive T cells, specific induc- [54].
tion of Tregs and switch in the Th1/Th2 balance in favour of Th2
[49,50]. 5.2. Engineering lymphocytes
Although promising results have been obtained in several
mouse models, their translation to humans is highly challenging, As previously mentioned, pathological lymphocytes recognising
mainly because of the costs and the wide range of available methods self-components are present in RA patients. Autoreactive B cells are
to modulate DC functions and produce clinical grades of either allo- indeed major drivers of the pathology since B-cell deficient mice do
genic or autologous tolDCs. Three main phase I clinical trials have not develop CIA following immunisation [55]. Furthermore, B cell
been launched to test the efficacy of TolDC therapy in RA patients depletion by monoclonal antibodies has demonstrated strong pos-
(Table 1). itive impacts on disease courses in RA patients [56]. Conversely,
One study used autologous tolDCs differentiated ex vivo from B cells can exhibit tolerogenic properties, which may ameliorate
peripheral blood monocytes of RA patients, before loading with disease manifestations. For instance, one study also attempted to
four citrullinated self-peptides. RA patients then received a sin- take advantage of the natural antigen-presenting capacity of B cells.
gle intradermal injection of this compound (called “rheumavax”) By transducing hematopoietic stem cells before engraftment with
[51]. Rheumavax was well tolerated and appeared to be safe [51]. a lentiviral vector encoding a collagen II inside the CLIP peptide,
In addition, after one month of treatment, the ratio of Tregs to Teff presentation of collagen II epitopes on MHC-II complexes was com-
cells was significantly improved and the serum levels of certain pelled. This presentation produced a tolerogenic effect and reduced
pathological cytokines and chemokines were decreased in patients the incidence and severity of the pathology [57].
[51]. Alternatively, Tregs, as mentioned above, represent a subset of
Semi-mature autologous DCs pulsed with recombinant protein- T cells that exhibit immunosuppressive functions by suppressing
arginine deiminase type-4, heterogeneous nuclear ribonucleo- inflammation in an antigen-specific manner without impacting the
protein A2/B1, citrullinated filaggrin and citrullinated vimentin protective immune responses. Furthermore, Tregs have the ability
antigen have been alternately tested in RA patients [52]. The to home towards inflammatory sites, recognise their specific anti-
patients who received this cocktail (designated as CreaVax-RA) gens presented on APCs and locally suppress inflammation through
exhibited decreased numbers of interferon (IFN)-␥-producing T the secretion of cytokines or the expression of immunosuppress-
cells and autoantibody titers [52]. Of note, no clinical efficacy was ive surface molecules. Consequently, adoptive transfer of Tregs
observed for patients without any autoantibodies [52]. has been considered to restore tolerance and hence, to achieve

Table 1
Phase I clinical trials with TolDC to cure RA.

Trial Rheumavax CreaVax RA AuToDeCRA

Number of patients 29 12 13
DCs Autologous modified with a Autologous semi mature Autologous TolDCs
nuclear factor kB (NF-kB)
inhibitor
Antigen(s) Citrullinated peptides Recombinant PAD4, RA33, Synovial fluid antigens
citrullinated-filaggrin and
vimentin antigens
Number of infused cells Low dose: 7.2 × 103 to 1.7 × 104 Low dose: 0.5 × 107 Low dose: 1 × 106
High dose: 2.7 × 104 to High dose: 1.5 × 107 Medium dose: 3 × 106
6.2 × 104 High dose: 10 × 106
Number of injection(s) 1 5 1
Route of infusion Intradermal Subcutaneous Intra-articular
Monitoring 4 weeks to 6 months 14 and 24 weeks 1, 2 and 13 weeks
Clinical outcomes Improvement of the ratio Decrease of IFN-g producing Decrease of arthroscopic
Treg/Teff cells cells synovitis symptoms
Decrease of auto antibodies No systemic effects
Reference [51] [52] [53]

5
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

remission in several autoimmune diseases, including RA. Although pre-clinical observations. Several reasons might explain this dis-
adoptive transfer of non-specific or self peptide-specific Tregs has crepancy.
improved clinical symptoms in CIA, autoantigen specific Treg cell Firstly, disease-causing immunogenic self-antigens are often
transfer (Hsp60–specific CD8+ or collagen-II-specific type 1 Tregs) unknown for patients, while many arthritis mouse models rely
also mediated reduction of disease [58,59]. on the injection of a given self-peptide (mainly collagen II or PG),
The clinical efficacy of Tregs transfer has been hampered by the whose pathogenic role in humans remains unclear. In addition,
high amount of pro-inflammatory cytokines in the joints, which the appropriate self-peptide(s) for treatment would be different
could not be counterbalanced by the infused cells. Another lim- among patients. To address this issue, libraries of self-peptides
itation of Tregs lies in the difficulty to characterise and expand for treatment might become available not only for adapting the
these cells. Although some methods were developed to expand therapy to every patient, but also to restore the tolerance against
autoreactive Tregs, such as their exposure to autoantigen-loaded several self-peptides at the same time, thus counteracting epi-
APCs, producing high amount of antigen-specific Tregs remains a tope spreading. Additionally, by selecting organ-specific antigens
major challenge [60]. To circumvent the small number of autore- that are expressed in the joints for instance, tolerance to unknown
active Tregs that can be retrieved and expanded, Tregs may be disease-causing self-antigens might also be restored by bystander
genetically engineered in order to redirect their specificity against effect. Nevertheless, animal models that more closely mimic human
self-antigens. Accordingly, ectopic autoreactive T-cell receptor pathologies are required to achieve a deeper understanding of the
(TCR) or chimeric-antigen receptors (CAR) can be expressed after pathology and testing for the most appropriate therapeutic strat-
gene transfer in Tregs. Using this latter method, adequate numbers egy. Towards this goal, several mouse models humanised with
of auto-antigen specific TCR or CAR-engineered Tregs can easily either human joint molecules or cells, or with human immune cells
be manufactured. Paving the way for translation in autoimmune have been developed in the past decades and will certainly become
disorders, Fransson et al. expressed a CAR targeting myelin oligo- more widely used in preclinical studies on RA [63].
dendrocyte glycoprotein along with FoxP3 in T cells, which, after Secondly, the time course of monitoring of the clinical trials
intranasal application, could suppress an ongoing encephalomyeli- needs to be carefully considered and standardised, as the beneficial
tis [60]. effects might take several months or years to be detected, since the
Alternatively, T cells may also be engineered to directly suppress return to a state of tolerance is likely not immediate.
only pathologic B cells. Toward this goal, novel chimeric receptors, Finally, most studies in mice tested prophylactic approaches,
referred as BAR – for B-cell antibody receptor, have been gener- while in humans, the treatments generally start when the disease
ated by replacing the scFv of the CAR by the self-antigen itself [61]. is already well established, which limits their efficacy. Previ-
Promising results with transfer of BAR-engineered T cells leading ous or ongoing treatments of patients with anti-inflammatory
to inhibition of antigen-specific B cells in the context of hemophilia drugs may also impact the efficacy. Interestingly, autoantibodies
[62] indicate that BARs may also be used for RA. are often detected years before the onset of the disease, leaving
Overall, with the advances in proteomics, gene engineering a room for restoration of tolerance, before the disease reaches
and editing techniques, cell therapy with engineered lymphocytes an active phase. Further knowledge on prognostic biomarkers,
represents a very appealing approach to cure RA. Nevertheless, disease-causing agents and pathogenesis will obviously help for
important questions remain to be considered, such as the phys- the early disease management.
iological consequences of modified cell infusion, as well as the Alternatively, besides self-peptides, other molecules may be
long-term persistence of edited cells, their re-activation potential used as disease biomarkers, such as pro-inflammatory cytokines.
and their differentiation. For instance, a chimeric inflammation-sensitive promoter induc-
ing the expression of IL-10 in immune cells was shown to strongly
impact arthritis incidence and severity [64]. In addition, with the
6. Conclusion and perspectives expansion of molecular engineering, programmable synthetic sen-
sors can be designed in order to recognise a disease-biomarker
Because of their potentially life-long therapeutic effect, sev- molecule that can be chosen to trigger the expression of a desired
eral strategies to restore antigen-specific tolerance have emerged effector in response to sensing [65].
towards remission of RA. The first attempts relied on the oral inges- Overall, approaches to achieve antigen-specific tolerance hold
tion of self-peptides, with promising results in animal models. great promises for long-term RA treatment and will surely benefit
However, clinical trials failed to meet the expected improvement from technological progresses, such as gene and cell engineering, in
criteria of clinical scores. Self-peptides have been engineered to order to develop clinically safer and more effective strategies in the
enhance their intrinsic tolerogenic potential, either by modify- near future. Additionally, similar approaches may also be tested for
ing epitopes to render them more active immunologically, by other autoimmune pathologies and for overcoming organ rejection
combining several self-peptides or by conjugating immunosup- after transplant. Conversely, the development of efficient treat-
pressive drugs. Carriage of self-peptides on nanoparticles combined ments for other autoimmune pathologies would certainly widen
with cell-targeting molecules and immuno-modulatory drugs has the possibilities for RA treatment as well.
emerged as a potent platform to re-induce tolerance to self-
components.
More recently, transfer of immune cells has been considered as Author contributions
potential tolerogenic solutions for many immune-related diseases,
such as RA. For instance, tolerogenic dendritic cell differentiated A.P. conceptualised the manuscript, wrote the initial draft and
ex vivo and loaded with self-peptides was shown to efficiently revised subsequent drafts, F.F and F.-L.C. reviewed and edited the
reduce disease severity and incidence. Alternatively, the transfer manuscript prior to submission. All authors have read and agreed
of autoreactive Tregs, expanded ex vivo or genetically engineered to the published version of the manuscript.
to express self-antigen-specific chimeric receptors or self-peptides,
has emerged as a potential approach to cure RA.
Although promising results have been obtained in mouse Disclosure of interest
models, the translation to human pathologies remains highly chal-
lenging since the results of clinical trials often do not match the The authors declare that they have no competing interest.

6
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

Acknowledgments [24] Choy EH, Scott DL, Kingsley GH, et al. Control of rheumatoid arthritis by oral
tolerance. Arthritis Rheum 2001;44:1993–7 [https://doi.org/10.1002/1529-
0131(200109)44:9<1993::AID-ART347>3.0.CO;2-A].
The authors acknowledge the critical reading of this manuscript [25] Nagler-Anderson C, Bober LA, Robinson ME, et al. Suppression of
by Dr. Alexandre Belot. type II collagen-induced arthritis by intragastric administration
of soluble type II collagen. Proc Natl Acad Sci 1986;83:7443–6,
http://dx.doi.org/10.1073/pnas.83.19.7443.
[26] Jorgensen C, Gedon E, Jaquet C, et al. Gastric administration of recombinant 65
References kDa heat shock protein delays the severity of type II collagen induced arthritis
in mice. J Rheumatol 1998;25:763–7.
[1] Firestein GS, McInnes IB. Immunopathogenesis of rheumatoid arthritis. Immu- [27] Staines NA, Harper N, Ward FJ, et al. Mucosal tolerance and sup-
nity 2017;46:183–96, http://dx.doi.org/10.1016/j.immuni.2017.02.006. pression of collagen-induced arthritis (CIA) induced by nasal inhalation
[2] Wright JK, Cawston TE, Hazleman BL. Transforming growth factor beta stimu- of synthetic peptide 184-198 of bovine type II collagen (CII) express-
lates the production of the tissue inhibitor of metalloproteinases (TIMP) by ing a dominant T cell epitope. Clin Exp Immunol 2007;103:368–75,
human synovial and skin fibroblasts. Biochim Biophys Acta – Mol Cell Res http://dx.doi.org/10.1111/j.1365-2249.1996.tb08289.x.
1991;1094:207–10, http://dx.doi.org/10.1016/0167-4889(91)90010-U. [28] Garcia G, Komagata Y, Slavin AJ, et al. Suppression of collagen-induced
[3] Perricone C, Ceccarelli F, Saccucci M, et al. Porphyromonas gingi- arthritis by oral or nasal administration of type II collagen. J Autoimmun
valis and rheumatoid arthritis. Curr Opin Rheumatol 2019;31:517–24, 1999;13:315–24, http://dx.doi.org/10.1006/jaut.1999.0320.
http://dx.doi.org/10.1097/BOR.0000000000000638. [29] Honda A, Ametani A, Matsumoto T, et al. Vaccination with an immuno-
[4] Ishikawa Y, Terao C. The impact of cigarette smoking on dominant peptide of bovine type II collagen induces an anti-TCR response, and
risk of rheumatoid arthritis: a narrative review. Cells 2020:9, modulates the onset and severity of collagen-induced arthritis. Int Immunol
http://dx.doi.org/10.3390/cells9020475. 2004;16:737–45, http://dx.doi.org/10.1093/intimm/dxh075.
[5] Kerkman PF, Fabre E, van der Voort EIH, et al. Identification and char- [30] Miyahara H, Myers LK, Rosloniec EF, et al. Identification and characterisation of
acterisation of citrullinated antigen-specific B cells in peripheral blood a major tolerogenic T-cell epitope of type II collagen that suppresses arthritis
of patients with rheumatoid arthritis. Ann Rheum Dis 2016;75:1170–6, in B10.RIII mice. Immunology 1995;86:110–5.
http://dx.doi.org/10.1136/annrheumdis-2014-207182. [31] Ho DT, Wykoff-Clary S, Gross CS, et al. Growth inhibition of an
[6] Payet J, Goulvestre C, Bialé L, et al. Anticyclic citrullinated peptide established A431 xenograft tumour by a full-length anti-EGFR antibody
antibodies in rheumatoid and non-rheumatoid rheumatic disor- following gene delivery by AAV. Cancer Gene Ther 2009;16:184–94,
ders: experience with 1162 patients. J Rheumatol 2014;41:2395–402, http://dx.doi.org/10.1038/cgt.2008.68.
http://dx.doi.org/10.3899/jrheum.131375. [32] Xinqiang S, Fei L, Nan L, et al. Construction and characterisation of a
[7] Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classifica- novel DNA vaccine that is potent antigen-specific tolerising therapy for
tion criteria: an American College of Rheumatology/European League Against experimental arthritis by increasing CD4+ CD25+ Treg cells and inducing
Rheumatism collaborative initiative. Arthritis Rheum 2010;62:2569–81, Th1 to Th2 shift in both cells and cytokines. Vaccine 2009;27:690–700,
http://dx.doi.org/10.1002/art.27584. http://dx.doi.org/10.1016/j.vaccine.2008.11.090.
[8] Uysal H, Nandakumar KS, Kessel C, et al. Antibodies to citrullinated proteins: [33] Antonio López-Guerrero J, Ortiz MA, Páez E, et al. Therapeutic
molecular interactions and arthritogenicity. Immunol Rev 2010;233:9–33, effect of recombinant vaccinia virus expressing the 60-kd heat-
http://dx.doi.org/10.1111/j.0105-2896.2009.00853.x. shock protein on adjuvant arthritis. Arthritis Rheum 1994;37:1462–7,
[9] Darrah E, Andrade F. Rheumatoid arthritis and citrullination. http://dx.doi.org/10.1002/art.1780371009.
Curr Opin Rheumatol 2018;30:72–8, http://dx.doi.org/10.1097/ [34] Tang B, Cullins DL, Zhou J, et al. Modulation of collagen-induced arthritis by
BOR.0000000000000452. adenovirus-mediated intra-articular expression of modified collagen type II.
[10] Boissier M-C, Biton J, Semerano L, et al. Origins of rheumatoid arthritis. Joint Arthritis Res Ther 2010;12:R136, http://dx.doi.org/10.1186/ar3074.
Bone Spine 2020;87:301–6, http://dx.doi.org/10.1016/j.jbspin.2019.11.009. [35] Eneljung T, Tengvall S, Jirholt P, et al. Antigen-specific gene therapy after
[11] Snir O, Widhe M, Hermansson M, et al. Antibodies to several citrullinated anti- immunisation reduces the severity of collagen-induced arthritis. Clin Dev
gens are enriched in the joints of rheumatoid arthritis patients. Arthritis Rheum Immunol 2013;2013:1–11, http://dx.doi.org/10.1155/2013/345092.
2010;62:44–52, http://dx.doi.org/10.1002/art.25036. [36] Gjertsson I, Laurie KL, Devitt J, et al. Tolerance induction using lentiviral
[12] Burmester GR, Pope JE. Novel treatment strate- gene delivery delays onset and severity of collagen II arthritis. Mol Ther
gies in rheumatoid arthritis. Lancet 2017;389:2338–48, 2009;17:632–40, http://dx.doi.org/10.1038/mt.2009.299.
http://dx.doi.org/10.1016/S0140-6736(17)31491-5. [37] Tengvall S, Eneljung T, Jirholt P, et al. Gene therapy induces antigen-
[13] Boissier M-C, Assier E, Biton J, et al. Regulatory T cells specific tolerance in experimental collagen-induced arthritis. PLOS ONE
(Treg) in rheumatoid arthritis. Joint Bone Spine 2009;76:10–4, 2016;11:e0154630, http://dx.doi.org/10.1371/journal.pone.0154630.
http://dx.doi.org/10.1016/j.jbspin.2008.08.002. [38] Myers LK, Tang B, Rosioniec EF, et al. PART III. Autoimmunity: an altered peptide
[14] Min S-Y, Hwang S-Y, Park K-S, et al. Induction of IL-10-producing ligand of type II collagen suppresses autoimmune arthritis. Crit Rev Immunol
CD4+ CD25+ T cells in animal model of collagen-induced arthritis by 2007;27:345–56, http://dx.doi.org/10.1615/CritRevImmunol.v27.i4.40.
oral administration of type II collagen. Arthritis Res Ther 2004;6:R213, [39] Xi C, Tan L, Sun Y, et al. A novel recombinant peptide contain-
http://dx.doi.org/10.1186/ar1169. ing only two T-cell tolerance epitopes of chicken type II collagen
[15] Friedman A, Weiner HL. Induction of anergy or active suppression follow- that suppresses collagen-induced arthritis. Mol Immunol 2009;46:729–37,
ing oral tolerance is determined by antigen dosage. Proc Natl Acad Sci http://dx.doi.org/10.1016/j.molimm.2008.10.016.
1994;91:6688–92, http://dx.doi.org/10.1073/pnas.91.14.6688. [40] Gertel S, Serre G, Shoenfeld Y, et al. Immune tolerance induction with multi-
[16] Miller SD, Turley DM, Podojil JR. Antigen-specific tolerance strategies for epitope peptide derived from citrullinated autoantigens attenuates arthritis
the prevention and treatment of autoimmune disease. Nat Rev Immunol manifestations in adjuvant arthritis rats. J Immunol 2015;194:5674–80,
2007;7:665–77, http://dx.doi.org/10.1038/nri2153. http://dx.doi.org/10.4049/jimmunol.1402457.
[17] Zhu P, Li X-Y, Wang H-K, et al. Oral administration of type-II colla- [41] Spiering R, Margry B, Keijzer C, et al. DEC205+ dendritic cell-
gen peptide 250-270 suppresses specific cellular and humoral immune targeted tolerogenic vaccination promotes immune tolerance in
response in collagen-induced arthritis. Clin Immunol 2007;122:75–84, experimental autoimmune arthritis. J Immunol 2015;194:4804–13,
http://dx.doi.org/10.1016/j.clim.2006.08.004. http://dx.doi.org/10.4049/jimmunol.1400986.
[18] Khare SD, Krco CJ, Griffiths MM, et al. Oral administration of an immuno- [42] Zimmerman DH, Taylor P, Bendele A, et al. CEL-2000: a therapeutic vac-
dominant human collagen peptide modulates collagen-induced arthritis. J cine for rheumatoid arthritis arrests disease development and alters serum
Immunol 1995;155:3653–9. cytokine/chemokine patterns in the bovine collagen type II induced arthri-
[19] Trentham DE. Evidence that type II collagen feeding can induce a durable ther- tis in the DBA mouse model. Int Immunopharmacol 2010;10:412–21,
apeutic response in some patients with rheumatoid arthritis. Ann N Y Acad Sci http://dx.doi.org/10.1016/j.intimp.2009.12.016.
1996;778:306–14, http://dx.doi.org/10.1111/j.1749-6632.1996.tb21138.x. [43] Kim E-K, Seo H-S, Chae M-J, et al. Enhanced anti-tumour immunothera-
[20] Trentham D, Dynesius-Trentham R, Orav E, et al. Effects of oral administra- peutic effect of B-cell-based vaccine transduced with modified adenoviral
tion of type II collagen on rheumatoid arthritis. Science 1993;261:1727–30, vector containing type 35 fibre structures. Gene Ther 2014;21:106–14,
http://dx.doi.org/10.1126/science.8378772. http://dx.doi.org/10.1038/gt.2013.65.
[21] Sieper J, Kary S, Sörensen H, et al. Oral type II collagen treatment in early [44] Capini C, Jaturanpinyo M, Chang H-I, et al. Antigen-specific suppression
rheumatoid arthritis. A double-blind, placebo-controlled, randomised trial. of inflammatory arthritis using liposomes. J Immunol 2009;182:3556–65,
Arthritis Rheum 1996;39:41–51, http://dx.doi.org/10.1002/art.1780390106. http://dx.doi.org/10.4049/jimmunol.0802972.
[22] Wei W, Zhang L-L, Xu J-H, et al. A multicentre, double-blind, randomised, con- [45] Galea R, Nel HJ, Talekar M, et al. PD-L1 and calcitriol-dependent liposomal
trolled phase III clinical trial of chicken type II collagen in rheumatoid arthritis. antigen-specific regulation of systemic inflammatory autoimmune disease. JCI
Arthritis Res Ther 2009;11:R180, http://dx.doi.org/10.1186/ar2870. Insight 2019;4:e126025, http://dx.doi.org/10.1172/jci.insight.126025.
[23] Barnett ML, Kremer JM, St Clair EW, et al. Treatment of [46] Bednar KJ, Nycholat CM, Rao TS, et al. Exploiting CD22 to selectively tolerise
rheumatoid arthritis with oral type II collagen. Results of a mul- autoantibody producing B-cells in rheumatoid arthritis. ACS Chem Biol
ticentre, double-blind, placebo-controlled trial. Arthritis Rheum 2019;14:644–54, http://dx.doi.org/10.1021/acschembio.8b01018.
1998;41:290–7 [https://doi.org/10.1002/1529-0131(199802)41:2<290::AID- [47] Pozsgay J, Babos F, Uray K, et al. In vitro eradication of citrulli-
ART13>3.0.CO;2-R]. nated protein specific B-lymphocytes of rheumatoid arthritis patients

7
A. Page, F. Fusil and F.-L. Cosset Joint Bone Spine 88 (2021) 105164

by targeted bifunctional nanoparticles. Arthritis Res Ther 2016;18:15, [57] Andersson SEM, Eneljung T, Tengvall S, et al. Collagen epitope expression on B
http://dx.doi.org/10.1186/s13075-016-0918-0. cells is sufficient to confer tolerance to collagen-induced arthritis. Arthritis Res
[48] Hilkens CMU, Isaacs JD. Tolerogenic dendritic cell therapy for rheumatoid Ther 2016;18:140, http://dx.doi.org/10.1186/s13075-016-1037-7.
arthritis: where are we now?: current status of tolDC therapy. Clin Exp [58] Asnagli H, Martire D, Belmonte N, et al. Type 1 regulatory T cells specific for
Immunol 2013;172:148–57, http://dx.doi.org/10.1111/cei.12038. collagen type II as an efficient cell-based therapy in arthritis. Arthritis Res Ther
[49] Jansen MAA, Spiering R, Ludwig IS, et al. Matured tolerogenic dendritic cells 2014;16:R115, http://dx.doi.org/10.1186/ar4567.
effectively inhibit autoantigen specific CD4+ T cells in a murine arthritis model. [59] Leavenworth JW, Tang X, Kim H-J, et al. Amelioration of arthritis through
Front Immunol 2019;10:2068, http://dx.doi.org/10.3389/fimmu.2019.02068. mobilisation of peptide-specific CD8+ regulatory T cells. J Clin Invest
[50] Ning B, Wei J, Zhang A, et al. Antigen-specific tolerogenic dendritic cells 2013;123:1382–9, http://dx.doi.org/10.1172/JCI66938.
ameliorate the severity of murine collagen-induced arthritis. PLOS ONE [60] Fransson M, Piras E, Burman J, et al. CAR/FoxP3-engineered T regulatory cells
2015;10:e0131152, http://dx.doi.org/10.1371/journal.pone.0131152. target the CNS and suppress EAE upon intranasal delivery. J Neuroinflammation
[51] Benham H, Nel HJ, Law SC, et al. Citrullinated peptide den- 2012;9:576, http://dx.doi.org/10.1186/1742-2094-9-112.
dritic cell immunotherapy in HLA risk genotype-positive [61] Ellebrecht CT, Bhoj VG, Nace A, et al. Reengineering chimeric antigen receptor
rheumatoid arthritis patients. Sci Transl Med 2015;7:290ra87, T cells for targeted therapy of autoimmune disease. Science 2016;353:179–84,
http://dx.doi.org/10.1126/scitranslmed.aaa9301. http://dx.doi.org/10.1126/science.aaf6756.
[52] Joo YB, chan-Bum C, Jin-ah J, et al. Phase 1 study of immunotherapy using [62] Parvathaneni K, Zhang A, Kim YC, et al. BAR-CD8 T-cell mediated targeted
autoantigen- loaded dendritic cells in patients with anti-citrullinated peptide killing of inhibitor producing FVIII-specific B cells. Blood 2015;126:294,
antigen positive rheumatoid arthritis; 2014. http://dx.doi.org/10.1182/blood.V126.23.294.294.
[53] Bell GM, Anderson AE, Diboll J, et al. Autologous tolerogenic dendritic cells [63] Schinnerling K, Rosas C, Soto L, et al. Humanised mouse models
for rheumatoid and inflammatory arthritis. Ann Rheum Dis 2017;76:227–34, of rheumatoid arthritis for studies on immunopathogenesis and pre-
http://dx.doi.org/10.1136/annrheumdis-2015-208456. clinical testing of cell-based therapies. Front Immunol 2019;10:203,
[54] Park J-E, Jang J, Choi J-H, et al. DC-based immunotherapy combined with http://dx.doi.org/10.3389/fimmu.2019.00203.
low-dose methotrexate effective in the treatment of advanced CIA in mice. [64] Henningsson L, Eneljung T, Jirholt P, et al. Disease-dependent local IL-
J Immunol Res 2015;2015:1–15, http://dx.doi.org/10.1155/2015/834085. 10 production ameliorates collagen-induced arthritis in mice. PLoS ONE
[55] Svensson L, Jirholt J, Holmdahl R, et al. B cell-deficient mice do not develop 2012;7:e49731, http://dx.doi.org/10.1371/journal.pone.0049731.
type II collagen-induced arthritis (CIA). Clin Exp Immunol 1998;111:521–6, [65] Kojima R, Aubel D, Fussenegger M. Building sophisticated sensors of extracellu-
http://dx.doi.org/10.1046/j.1365-2249.1998.00529.x. lar cues that enable mammalian cells to work as “doctors” in the body. Cell Mol
[56] Mok CC. Rituximab for the treatment of rheumatoid arthritis: an update. Drug Life Sci 2020;77:3567–81, http://dx.doi.org/10.1007/s00018-020-03486-y.
Des Dev Ther 2013;87, http://dx.doi.org/10.2147/DDDT.S41645.

You might also like