T cells
The second main player in the field of systemic sclerosis is the T cell. T cells have been shown to modulate and drive the development of autoimmunity, inflammatory response and fibrosis. In the course of disease, alteration of the frequencies of lymphocytes is observed suggesting the pivotal role of these cells in progression of the disease [
28]. The role of T cells in a pathophysiological context in systemic sclerosis is recognized as T cells interact with B cells to produce specific autoantibodies as well as a source of potent proinflammatory cytokines [
29]. T helper (Th) cells are involved both in the early and late phase of the disease. Taking into consideration the concept of Th cells polarisation toward Th1 or Th2 response is worth to note that Th1 but also Th17 related cytokines as TNFα, IFN, IL-1,IL-12, IL17 act predominantly at early stages of the disease being responsible for driving the inflammation [
30]. With progression of the disease inflammation is less active and Th2 cytokines (IL-4, IL-13, IL-5, IL-6, and IL-10) become more prominent [
31,
32].
Modulation of T cell function may also be realized indirectly by interaction with cell-to cell signalling (co-stimulation). Essential molecules involved in this process are the inducible T cell costimulator ICOS and the CD 28 molecule that bind ICOS ligand and CD80/86 ligands respectively. Acazicolcept a double ICOS and CD28 antagonist has been tested in a mouse model of systemic sclerosis. Treatment with Acazicolcept induced a significant reduction in dermal thickness, collagen content, myofibroblast number, and immunocompetent cells number (B cells, T cells, neutrophils, and macrophages) in the Fra-2 Tg mouse model. Additionally acazicolcept treatment reduced lung fibrosis and right ventricular systolic pressure (RVSP). Moreover treatment with acazicolcept resulted in reduction of frequency of CD4+ and T effector memory cells and an increase in the percentage of CD4+ T naïve cells in spleen and lung animals treated [
33].
The other therapeutic option is targeting CD30. The CD30 molecule, also known as Ki-1 or TNFRSF8, was first identified in 1982 in patients with Hodgkin (HL) lymphoma. The expression of this molecule is however not restricted to Hodgkin and Reed Stenberg cells. CD30 is expressed on a small subset of activated T and B lymphocytes, and a variety of lymphoid neoplasms, with the highest expression in classical HL and anaplastic large cell lymphoma. Subsequent studies demonstrated that CD30 expression was not uniform across all activated lymphocytes, instead being limited to subpopulations of CD4
+/CD45RO
+and CD8
+T cells in lymph nodes and the thymic medulla. CD30 expression appears higher in CD4+ and CD8+ cells producing a Th2-type cytokines [
34,
35,
36]. Soluble CD30 molecule has been found in high concentrations in sera of patients with systemic sclerosis that indirectly suggests the pathogenic role of this molecule and in wider context the Th2 response in the development of the disease [
37,
38,
39,
40,
41,
42]. At the moment two clinical trials are ongoing to test the safety and efficacy of Brentuximab vedotin (BV), a CD30-directed antibody-drug conjugate, which is already approved by US FDA for treatment of classic Hodgkin lymphoma (cHL). The first results in patients with SSc will be available in 2023 (NCT03222492 and NTC03198689).
Full activation of T cells is a multi-step process that requires not only the recognition of antigens by T cells but, also a second co-stimulatory signal, provided by binding of the CD28 receptor on T cells to CD80 and/or CD86 molecules on the surface of antigen-presenting cells. [
43]. At the time of T cell activation the CD4 cell may express a regulatory molecule namely CD152 or Cytotoxic T lymphocyte-associated protein-4 (CTLA-4), an inhibitory receptor expressed constitutively on CD4+CD25+ T regulatory lymphocytes (Treg) and transiently on activated CD4+ and CD8+ T lymphocytes. With very high affinity to CD28 (150 times more than CD80/86), CD152 is able to halt activation of T cells preventing an uncontrolled immune response [
44]. That was a theoretical background for synthesis and introduction of the fusion protein composed from a soluble form of the extracellular domain of CTLA-4 linked to immunoglobulin G1Fc part (Abatacept, Orencia) [
45]. It showed its safety and efficacy in rheumatoid arthritis and is still intensively tested in various autoimmune diseases [
46,
47,
48,
49,
50,
51,
52,
53]. In systemic sclerosis, which is characterised by a pivotal role of T cell orchestrating immune response [
54] blocking the T cell activation seems to be a reasonable approach. Indeed, data from a small retrospective multicentre study with 27 patients showed the potential utility of abatacept in the treatment of systemic sclerosis, where reduction of skin involvement as well as improvement in tender and swollen join count were observed [
55]. Following these promising data the ASSET trial (A Study of Subcutaneous Abatacept to Treat Diffuse Cutaneous Systemic Sclerosis), which is a phase 2, double-blind, placebo-controlled trial of weekly subcutaneous abatacept over 12 months in patients with early diffuse cutaneous systemic sclerosis (≤36 months of disease) has been started .The ASSET trial showed only moderate (statistically non-significant) improvement with abatacept in the primary endpoint of mean change from baseline to month 12 in skin score [
56]. An open label extension phase of this study confirmed the good safety profile of abatacept, however again the primary endpoint of change in mRSS has not been reached [
57]. In spite of not reaching a significant improvement in skin score abatacept remains a potential candidate as a disease modifying drug in systemic sclerosis and more trials are needed to confirm the role of this drug in the treatment of the disease.
Targeting the specific cytokines
Skin fibrosis, vasculopathy and inflammation are three crucial elements of disease background. Each process is precisely orchestrated by cytokines, and chemokines activity, thus the rationale approach is to block or (rarely) enhance function of a given cytokine. Among many cytokines already identified only few of them participate in the beginning and the progression of SSc.Keeping in mind many limitations of pure categorisation of autoimmune diseases as Th1 or Th2 dependent condition. SSc may be categorised as at least partially,Th2 dependent disease with the prominent Th2 immune response and the subsequent release of Th2-dependent cytokines such as IL-4, IL-5, and IL-13, which are able to control the fibrotic process [
58].
In the early stages of scleroderma however, Th1 cells and Th17 cells are suggested to dominate the immune profile [
59] later shifting to Th2 [
60] and this may explain why patients with SSc are characterized by overexpression of IFNα at the early stages of the disease, which than translates to the disease’s development [
61,
62]. It shows clearly that both arms of the immune response – cellular and humoral are involved into pathogenesis of SSc however their role depends on the stage of the disease. This may bring several therapeutic consequences suggesting that direct therapeutic targeting of a cytokine should be done in the proper phase of the disease.
Several studies have shown that IL-4, a typical representative of Th2 dependent response plays an important role as a profibrotic cytokine that stimulates collagen synthesis by fibroblasts. This was proven in laboratory studies where IL-4
stimulated human dermal fibroblasts to synthesize types I and III collagen and fibronectin [
63,
64]. High levels of IL-4 were reported in patients with systemic sclerosis, where it plays a pathogenetic role inducing the formation of the extracellular matrix [
65,
66].
Even more studies refer to the other typical profibrotic cytokine IL-13. IL-13 has several similarities to IL-4 at the amino acid level and both cytokines display 20-30% homology [
67]. Moreover they show similar biological activity since they signal through receptor heterodimers built with combinations of three possible subunits: IL-4Rα, the common gamma chain (γc), and IL-13Rα1 common receptor chain [
68]. In detail, IL-4 and IL-13 share common type I cytokine receptor composed with IL-4α and gamma chain (γc) receptor alpha (IL-4Rα), coupled with the Janus kinase/signal transducer and activator of transcription protein 6 (JAK/STAT6) signaling pathway.
The other receptor, IL-13 may bind to is the IL-13Rα2. IL-13 preferentially binds to this type of the receptor with very high affinity. Binding of the cytokine however does not exert any physiological response as this type of receptor is commonly considered a “decoy” receptor as it has a short cytoplasmic tail with no recognizable signalling motifs [
69]. Finally IL-4 and Il-13 may signal through type II cytokine receptor composed with IL-4Rα and Il-13Rα2. IL-4 and Il-13 are commonly recognized as leading cytokines driving the fibrotic process [
70,
71]. Keeping in mind that both cytokines signal through type I or type II cytokine receptor attached to protein kinases, commonly referred as Janus kinase, with subsequent activation of JAK/STAT pathway and finally to control several gene expression, resulting in cytokine synthesis and escalation of the fibrotic process [
72,
73]. This finding may potentially open new therapeutic approaches. Type I and II cytokines attached to Janus kinases may be simply blocked with the use of small synthetic compounds – Janus kinase inhibitors [
74].With large profibrotic potential both IL-4 and IL-13 are natural targets to inhibit, aiming to ameliorate the fibrotic process [
75,
76]. Indeed, targeting IL-4, and IL-13 has been tested in various fibrotic diseases including systemic sclerosis [
70]. The main concept of targeting IL-4/IL-13 is based on the fact that these cytokines represent a Th2 response. The reasonable approach for SSc treatment is therefore to shift the immune response toward Th1. The IL-4, IL-13signalling pathway can be stopped at various levels: (1) by ameliorating the soluble cytokine activity, (2) targeting and inhibiting their receptors on cell surfaces or (3) blocking the intracellular signalling pathways. The first approach may be easily done by simply blocking IL-4/IL-13 activity with the use of monoclonal antibodies targeting either IL-4 or IL-13 activity. Quite recently data on a randomised, double-blind, placebo-controlled, 24-week, phase II, proof-of-concept study of romilkimab (SAR156597) in early diffuse cutaneous systemic sclerosis became available. In the study romilkimab, a humanised, bispecific immunoglobulin-G4 antibody that binds and neutralises IL-4/IL-13 has been tested in patients with diffuse type of SSc. In this study romilkimab demonstrated a significant effect on the skin changes in early dsSSc [
77]. However the results of the study should be interpreted cautiously as they referred to a relatively small group of patients that obviously requires confirmation in a future phase III study.
The second possibility is to block the receptor function with the use of a monoclonal antibody. As far as this approach is concerned dupilumab, a monoclonal humanized antibody targeting subunit IL-4Rα, recently commercialized for atopic dermatitis and asthma seems to be a reasonable approach [
78,
79]. At the moment however dupilumab has not been tested in systemic sclerosis.
The last approach is to block the signalling pathway with the use of JAK inhibitors [
74]. Cytokines are small molecular weight transmitters essential in cell to cell interaction to modulate innate and acquired immune response. Based on the similar structure they are typically divided into several cytokines families. Cytokines signal via typical receptors which can be categorized into several receptor superfamilies. Cytokines interact with the extracellular domain of the receptor, which when activated can transmit the signal via long chains of transmission molecules to activate specific genes in the nucleus. Among them cytokines belonging to class I and class II receptors family utilize receptors which lack intracellular enzymatic domain and require specific kinases to fully activate the receptor and enable to transmit the signal into the nucleus [
80]. The Janus kinase (JAK) family enzymes are non-receptor tyrosine kinases that phosphorylate cytokine receptors enabling them to start signalling through the JAK-STAT signalling pathway. Considering that JAK-STAT signal transduction is initiated by the binding of ligands, such as cytokines to their receptors, proper JAK activity in the JAK-STAT pathway is a key element to orchestrate immune response and dysfunctional JAKs are directly responsible for cancers, immune system-related diseases, and autoimmune disorders [
81]. These enzymes referred as Janus kinases or simply JAK kinases are an essential element for the transmission of cytokine signals for such important cytokines as IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 which transmit their signals via gamma chain receptors (γc) [
82], since GM-CSF, IL-3, and IL-5 signalling is via the beta type of receptor [
83]. Another class of receptor is the heterodimer composed of the gp 130 protein (or its homologue) which is responsible for signalling from IL- 6, IL-11, IL-31, IL-35, and IL-27 [
84]. Finally, the class I receptor subfamily which has a common p40 subunit interacts with IL-12 and IL-23 [
85]. The second group of receptors called class II receptors is responsible for transmitting signals from interferons and the IL-10 cytokine family (IL-10, IL-19, IL-20, IL-22, IL-24, and IL-26) [
86].
All types of receptor utilize a combination of the four known JAK kinases (JAK1, JAK2, JAK3 and TYK2). This makes it possible to stop or at least modulate cytokine signalling by blocking JAK activity. This was a theoretical background for the development of JAK inhibitors. Contrary to the large and high molecular weight biologics targeting one specific cytokine, JAK inhibitors are small synthetic compounds easy to synthetize and handle. This group of cytokine modulators showed their safety and efficacy in several rheumatological, dermatological and haematological conditions and are intensively tested in many others including connective tissue diseases and the other autoimmune disorders [
87,
88,
89,
90,
91].
The role of JAK kinase inhibition in SSc patients can be explained again in the context of the activity of the typical Th2-dependent cytokines IL-4 and IL-13, which are responsible for driving the profibrotic process in the body. It is worth noticing however, that blocking one JAK may also result in the inhibition of other potentially proinflammatory and profibrotic cytokines, so creating a strong antifibrotic milieu.
The other players in this field are the IL-12 family of cytokines. Since the disease is characterized by the activation of IL-12 cytokine family members responsible for driving the profibrotic effect, JAK inhibition signalling from IL-12 specific receptors may block IL-12, IL-23 and IL-27 thus ameliorating a significant profibrotic effect. The role of the IL-6 cytokine was already mentioned in the light of biologics (tocilizumab) tested in SSc. IL-6 activity amelioration may alternatively be blocked by JAK inhibitors. Again, with a single compound we are to block all IL-6 family members simply due to the similarities in IL-6 receptor family, where all types of IL-6 receptors are coupled with the JAK kinases.
The role of blocking a cytokine class II receptor in systemic sclerosis is less clear. From a theoretical point of view class II cytokines (IL-10 family cytokines and interferons) are able to transmit both pro and anti-inflammatory signals. In detail IL-10 is mainly produced by Bregs that was able to suppress skin fibrosis [
92]. It has also been shown that IL-10–producing Bregs have been found to be reduced in patients with SSc and correlated with disease activity, suggesting the anti-inflammatory potential of IL-10 in the disease [
93,
94]. Less is known on the role of the other IL-10 family cytokines such as IL-20 or IL-23 in the development of systemic sclerosis. Data from the literature suggests the reduced expression of IL-20 or dysregulated IL-23 signalling [
92,
95]. It remains an open question whether these findings are clinically important. In line with this, it is not clear if blocking this part of cytokine signalling may benefit patients with systemic sclerosis.
The therapeutic potential of JAKi may also be explained in another way. Systemic sclerosis is characterized by overexpression of interferon type 1 [
61,
96,
97]. This pathophysiological phenomenon commonly referred to as an interferon signature is characterized by overexpression of several proinflammatory – Interferon- related genes. Type 1 of interferons comprises a group of five classes of interferons namely: α, β, ω, ε and κ. All types of INT T1 classes signal through the same type 1 IFN heterodimeric receptor complex coupled with the JAK kinases. This brings novel therapeutic possibilities as blocking JAK with its inhibitor can block the whole interferons’ signalling. Blocking the interferon signalling seems to be relatively safe. According to a phase I study, administration of MEDI-546, ahuman immunoglobulin G1 kappa monoclonal antibody directed against IFNAR1, resulted in sustained inhibition of the type I IFN gene signature [
98]. Followed this promising results the study with Anifrolumab (anti-IFNAR1 monoclonal antibody) showed the reduced suppression of the IFN signature and TGFβ signalling in SSc skin [
99]. This shows that blocking the IFN receptor might be a promising way to reduce disease activity in subjects with SSc or other IFN-related inflammatory diseases. Whether this finding translates to the inhibition of IFN signalling via JAK inhibitors should be elucidated.
Recent analysis of case reports and small uncontrolled trials showed the clinical benefit of JAK inhibitors in interstitial lung diseases associated with SSc. Keeping in mind many limitations from these studies, the results may indicate that targeting several cytokines with one drug may be a promising way to halt the progression of fibrosis and inflammation [
100].