Next Article in Journal
Neutrophil Extracellular Vesicles and Airway Smooth Muscle Proliferation in the Natural Model of Severe Asthma in Horses
Next Article in Special Issue
Hematopoietic Stem Cell Transplantation in Refractory Crohn’s Disease: Should It Be Considered?
Previous Article in Journal
Biological Roles of Fibroblasts in Periodontal Diseases
Previous Article in Special Issue
Mesenchymal Stem Cell-Derived Secretome: A Potential Therapeutic Option for Autoimmune and Immune-Mediated Inflammatory Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches

by
Elisabetta Xue
1,*,†,
Antonina Minniti
2,*,†,
Tobias Alexander
3,4,
Nicoletta Del Papa
2,‡,
Raffaella Greco
1,‡ and
on behalf of The Autoimmune Diseases Working Party (ADWP) of the European Society for Blood and Marrow Transplantation (EBMT)
1
Hematopoietic and Bone Marrow Transplant Unit, San Raffaele Hospital, 20132 Milan, Italy
2
Department of Rheumatology, ASST G. Pini-CTO, 20122 Milan, Italy
3
Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
4
Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117 Berlin, Germany
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cells 2022, 11(21), 3346; https://doi.org/10.3390/cells11213346
Submission received: 24 September 2022 / Revised: 19 October 2022 / Accepted: 19 October 2022 / Published: 24 October 2022
(This article belongs to the Special Issue Stem Cell Therapy for Autoimmune Diseases)

Abstract

:
Systemic sclerosis (SSc) is a systemic disease characterized by autoimmune responses, vasculopathy and tissue fibrosis. The pathogenic mechanisms involve a wide range of cells and soluble factors. The complexity of interactions leads to heterogeneous clinical features in terms of the extent, severity, and rate of progression of skin fibrosis and internal organ involvement. Available disease-modifying drugs have only modest effects on halting disease progression and may be associated with significant side effects. Therefore, cellular therapies have been developed aiming at the restoration of immunologic self-tolerance in order to provide durable remissions or to foster tissue regeneration. Currently, SSc is recommended as the ‘standard indication’ for autologous hematopoietic stem cell transplantation by the European Society for Blood and Marrow Transplantation. This review provides an overview on cellular therapies in SSc, from pre-clinical models to clinical applications, opening towards more advanced cellular therapies, such as mesenchymal stem cells, regulatory T cells and potentially CAR-T-cell therapies.

1. Introduction

Systemic sclerosis (SSc) is a rare autoimmune systemic disease characterized by three main dysfunctions: autoimmune responses, vasculopathy and tissue fibrosis. The result is a progressive loss of the microvascular bed and the development of fibrosis of the skin and internal organs, including lung and gastrointestinal tract [1]. The first and most frequent symptom of presentation is Raynaud’s phenomenon, a direct sign of vascular dysfunction often complicated by digital ulcers (DU), one of the major burdens for SSc patients [2]. Another manifestation of vasculopathy is pulmonary arterial hypertension which, along with extensive skin and internal organ fibrosis, is responsible for increased disability and mortality in SSc patients [3,4].
The etiopathogenesis of the disease is complex, but substantial progress has been made in understanding the mechanisms involved. It is assumed that, in a genetically predisposed population, environmental factors lead to cytokines release, immune cell activation and connective tissue repair dysregulation [5,6,7]. Indeed, the fibrotic changes observed in SSc patients are likely due to a dysfunctional repair of connective tissues in response to injuries like oxidative stress. A wide range of cellular components are involved, mainly endothelial cells, mesenchymal cells, and T and B lymphocytes. Fibrosis is caused by excessive collagen and other extracellular matrix proteins’ production by myofibroblasts. The transition of endothelial cells to myofibroblast is induced by cytokines released by activated B and T cells (IL-6, IFN-alfa, IL-10, TGF-beta, PDGF) [8].
Currently, SSc is considered as incurable. The recent introduction of biologic disease-modifying drugs (DMARDs) and anti-fibrotic therapies has provided more specificity and effectiveness. However, such therapies usually only slow the disease progression and rarely reverse disease manifestations. Moreover, maintenance therapies expose patients to side effects, such as infections, and may be associated with a cumulative risk of co-morbidities [9]. The need to reset autoimmune pathogenic mechanisms before the appearance of extensive and irreversible damage has led to many efforts to develop cell-based therapies able to restore self-tolerance, from animal models to clinical practice [10]. Along with their tolerogenic properties, cell-based therapies showed an interesting potential as regenerative therapies able to repair tissues already damaged.
Here, we review the available literature on cell-based therapies in SSc, from pre-clinical models to clinical applications, and approaches adopted in other autoimmune diseases potentially of interest for future investigations in SSc patients.

2. Hematopoietic Stem Cell Transplantation

Hematopoietic stem cell transplantation (HSCT) has a consolidate role in treating hematologic malignancies. For more than 20 years, there has been a growing interest in its application for the treatment of autoimmune diseases, and as a consequence of three positive RCT, autologous HSCT is now considered a standard therapy in refractory SSc [11,12]. HSCT is a multistep procedure designed to replace the hematopoietic system of a patient with a new one derived from HSCs, which can be collected from a healthy donor (allogeneic HSCT) or from the same patient (autologous). The graft can undergo CD34 selection in order to enrich HSCs and potentially remove self-reactive lymphocytes or can be infused unmanipulated. The ablation of autoreactive immune cells is achieved through a conditioning regimen, which can be highly variable in intensity, ranging from fully to non-myeloablative, and generally includes anti-thymocyte globulin (ATG). A myeloablative regimen can be busulfan- or total body irradiation-based, whereas non-myeloablative regimen are mostly cyclophosphamide- or fludarabine-based. Intermediate intensity regimens are also adopted, especially in neurological autoimmune diseases [13,14,15,16,17].
The rationale of autologous HSCT for autoimmune diseases is to reset a dysfunctional, autoreactive immune system with the intent to restore a more naïve, self-tolerant one [18,19]. Several mechanisms that might participate in immunologic reset have been described, including thymic reactivation [20,21] with a vast diversification of the TCR repertoire [21], expansion of regulatory T cells, [22,23] and restoration of a naïve B-cell compartment with re-emergence of regulatory B cells [21].
Among initial reports in SSc, [24,25] Farge and colleagues documented the safety and clinical benefit of autologous HSCT in 12 patients within a phase I-II study. Hematopoietic recovery was observed in all patients, and 8 out of 11 evaluable patients achieved significant clinical response [26]. After these promising initial results, three randomized–controlled studies have been conducted for SSc, comparing autologous HSCT to standard immunosuppressive therapies. The phase II ASSIST trial randomized nineteen SSc patients to receive either non-myeloablative, unmanipulated HSCT or monthly cyclophosphamide. All patients in the HSCT arm showed both cutaneous and pulmonary improvements, compared to lack of significant long-term benefit after standard immunosuppression with cyclophosphamide. Notably, the authors suggested that interstitial lung disease, long considered as being irreversible, might be at least partially reversed after HSCT, with sustained improvements observed years after transplant [15]. In the phase III ASTIS trial, 156 patients were randomized to receive either non-myeloablative, CD34-selected HSCT or standard immunosuppression with monthly cyclophosphamide pulse. Despite higher early treatment-related mortality in the HSCT arm (10% versus none, mainly attributed to SSc-related cardiac dysfunction), event-free survival and overall survival was superior to cyclophosphamide during follow-up [16]. Finally, in the SCOT trial, 75 patients received either myeloablative, total body irradiation-based CD34-selected HSCT or monthly cyclophosphamide pulses. In this study, there was remarkably low treatment-related mortality in the HSCT group, which might be explained by more stringent inclusion criteria at patient’s selection. Patients in the HSCT arm had a higher risk of severe infections in the early stages, but after a median of 54 months of follow up, the HSCT-group showed significant benefit in term of clinical outcomes compared to the control group [14]. HSCT effects in SSc patients were also evaluated at a molecular level by Assassi and colleagues comparing whole blood transcript and serum protein levels between patients in the HSCT arm and in the cyclophosphamide arm of the SCOT trial. The authors focused on a specific molecular signature of SSc, including high interferon level, high neutrophil gene expression profile and low cytotoxic/NK profile, and reported a significant amelioration of all parameters in the HSCT group, suggesting that HSCT may “correct” SSc-related dysfunction at a deeper level as compared to cyclophosphamide group. Interestingly, these molecular changes reflected improvement in pulmonary and skin involvement [27].
All these studies impressively demonstrated that sustained clinical improvements over years after HSCT are achievable, which led autologous HSCT to be acknowledged as standard of care for refractory SSc. However, careful patient selection is crucial for identifying subjects who might benefit from HSCT without major complications. In this regard, severe cardiopulmonary dysfunction at the time of transplant was identified as a major risk for treatment-related mortality, suggesting that candidate screening through right heart catheterization, cardiac imaging, and pulmonary function testing is recommended, and that a patient-tailored treatment should be adopted in high-risk categories [28,29].
More recently, results from a non-interventional prospective study with 80 included SSc patients demonstrated a 2-year progression-free survival of 81% after autologous HSCT. By multivariate analysis, higher baseline skin-modified Rodnan skin score and older age at transplant have been identified as predictors for lower progression-free survival, whereas graft manipulation CD34-selection was associated with superior responses [30]. The necessity of CD34 graft selection is controversial. Some studies reported improved clinical outcomes and better progression-free survival of CD34-selected transplants compared to unmanipulated transplants, with comparable toxicity and infection rate [30,31], while other failed to show any significant difference [32].
By May 2022, 776 patients with SSc have been treated with HSCT within the EBMT registry (Figure 1). While autologous HSCT is considered to be a standard treatment option [33], use of allogeneic HSCT for SSc remains anecdotal due to higher risk of transplant-related complications [34,35,36,37]. Reduced-toxicity HSCT platforms are needed to further investigate and expand allogeneic procedure in AD patients [38].

3. Adoptive Cellular Therapies

Alongside HSCT, other innovative therapeutic strategies with immunomodulatory properties have been investigated in the context of autoimmune disease, including SSc [12] (Figure 2).

4. Mesenchymal Stem Cells (MSC)

Mesenchymal stem cells, a heterogeneous population of stromal cells with high regenerative capacity that can be isolated, cultured, and expanded ex vivo, represent a promising source for cellular therapy approaches targeting SSc due to their immunosuppressive, angiogenic and anti-fibrotic properties [39]. Bone marrow (BM) stroma has been the main source for MSCs for decades; however, it became evident that MSCs with similar biological properties can also be isolated from placenta, Wharton’s jelly, blood vessels, dental pulp, derma and adipose tissue. The clinical interest in MSCs arises from their ability to differentiate toward mesodermal cell lineage, including chondroblasts, adipocytes and osteoblasts, and from their paracrine effects exerted through the secretion of trophic factors, cytokines, and other bioactive molecules within extracellular vesicles. MSCs plasticity and their regenerative and immunomodulatory properties are known to be driven by micro-environmental factors. This cellular plasticity reflects also on surfaces’ markers expression, which changes overtime due to senescence, inflammation, micro-environment changes and other pathological conditions. The International Society of Cellular Therapy (ISCT) has released guidelines regarding the minimum criteria that need to be met to define MSCs, which include multi-lineage differentiation potency, capacity to adhere to plastic surfaces, positive expression of CD105, CD73 and CD90 surface markers and negative expression of CD45, CD34, CD14, CD19 and HLA-DR surface markers [40].
MSCs of different tissue origin have been investigated for treating several indications, including tissue injuries, autoimmune diseases, and metastatic cancer [41,42,43,44,45,46]. Use of both autologous and allogeneic MSCs has been investigated. Given the low HLA expression on MSCs, there is not HLA-matching restriction when searching from MSC donors, minimizing the risk for immunological reactions [47]. However, recent studies reported some innate and adaptive immune responses triggered by allogeneic MSCs administration [48,49] despite being less potent compared to responses triggered by other cell lines.
Indeed, the destiny and long-term persistence of infused MSCs are a matter of debate. In preclinical models, syngeneic MSCs can persist for a long time, whereas allogeneic MSCs tend to be eliminated more rapidly [50]. Eggenhofer et al. observed viable MSCs only within the lungs after intravenous infusion, but not in other tissues [51]. Finally, monitoring of infused unmanipulated MSCs is challenging, due to a lack of cell-specific markers; a combination of cytokines’ hyper expression and inhibition above certain limits has been proposed as an indirect way to monitor MSCs potency and activity. In allogeneic setting, quantitative PCR chimerism can be potentially adopted for MSCs tracking.
Interestingly, the clinical effects of MSC-based therapy can often be observed for longer periods compared to their persistence in vivo. In a study on six patients with osteogenesis imperfecta treated with gene-marked MSCs, Horwitz et al. documented increased bone mineral density and reduced bone fractures despite only less than 2% of MSCs actually engrafted [52]. In an experimental model of SSc, Maria et al. showed how anti-fibrotic effect documented after MSCs infusion lasted up to 21 days, despite MSCs being undetectable after 7 days [53]. Similar results were shown in other settings, including myocardial infarction [54] and cerebral ischemia [55]; in the latter, MSCs injection determined prolonged benefits despite the majority of MSCs failing to engraft and only a minority differentiating into astrocytes.
This discrepancy has led to the hypothesis that MSCs’ biological effects do not rely only on cell engraftment and differentiation, but also on paracrine effects exerted through the secretion of trophic factors, cytokines and other bioactive molecules. For example, autologous MSCs collected from patients with SSc appear to increase levels of pro-angiogenic factors in vitro [56] and to promote revascularization in vivo [57] despite studies documenting their defective differentiation into endothelial cells [58]. In recent years, major interest has grown around MSCs-derived exosome vesicles, a relevant part of MSCs’ secretome that contain proteins, DNA, mRNA, and miRNA, which can be released in the extracellular environment and horizontally transferred to target cells [59]. Through these and other mechanisms, MSCs promote chemoattraction, cellular survival and growth, tissue repair. Furthermore, Bartholomew and colleagues initially reported that MSCs can influence immune system function by suppressing lymphocytes response and preventing in vivo rejection of skin graft. Subsequent experiments showed that MSCs shape the immune system by favoring the shift towards a regulatory T cell phenotype, inhibiting dendritic cell maturation and natural killer (NK) cells’ cytotoxicity, and downregulating B-cell proliferation. Notably, MSCs’ immunomodulatory properties depend on the inflammatory status, again confirming the susceptibility to the surrounding micro-environment. Indeed, some [60,61], but not all authors [62,63], reported that MSCs collected from patients with autoimmune diseases, despite showing similar differentiating potential, phenotype and surface markers expression compared to MSCs from healthy donors, display reduced clonogenic, proliferating and migrating capacity. A pro-fibrotic phenotype has also been reported in MSCs from these patients [64,65].
All these complex and fascinating characteristics have induced investigations towards MSCs as treatment for immune system dysregulation. A paradigmatic example is graft-versus-host disease [GvHD]. Since the first report of the successful use of third-party MSCs in a case of refractory acute GvHD [66], several phase I–II studies have shown the safety and applicability of this approach [67,68]. Despite some authors have raised concerns about higher infection rate [69] and relapse rate [70], subsequent studies did not confirm these observations. Remestemcel-L (Prochymal), an off-the-shelf BM-derived MSC product, has shown to be effective in combination with steroids as a first-line treatment for acute GvHD [71]; in the steroid-refractory GvHD setting, Remestemcel-L as a single agent led to 61% of overall response and significant improvement of survival outcomes [72]. MSCs role in GVHD prophylaxis is anecdotal, with controversial results; whereas some authors reported faster engraftment [73], decrease in acute GvHD rate [73,74,75] and chronic GvHD rate [76] in patients receiving prophylactic peri-transplant MSC infusion, others failed to observe any significant improvements [77,78]. Non-homogeneity of MSCs subtypes, dose and timing might partially explain these observations.

4.1. Intravenous MSCs Use

In animal models of SSc, several in vivo studies showed that the infusion of MSCs could limit the cellular damage and collagen deposition. For example, bone marrow-derived MSCs infused intravenously after exposure to bleomycin in a rat model of SSc resulted in decreased levels of tissue injury markers within the bronchoalveolar lavage fluid and decreased levels of proinflammatory cytokines [79]. Similarly, Moodley et al. reported improvement of pulmonary fibrosis after the infusion of umbilical cord-derived MSCs in a bleomycin-induced SSc model, with reduced levels of TGF-beta and IFN-gamma, as well as decreased collagen deposition within the lungs [80]. Subsequently, Yang et al. documented improvements in cutaneous fibrosis after the infusion of umbilical cord-derived MSCs in the a bleomycin-induced SSc model, with decreased collagen synthesis and inhibition of Th-17 cell function [81]. Analogous results have been obtained in the HOCI-induced model, in which the elevated levels of plasmatic nitric oxide and of cutaneous/lung tissue α-SMA and TGF-β1 normalized after umbilical cord- or bone marrow-derived MSC infusions, reaching nearly a normal histopathology of lung and skin [82,83,84] Interestingly, also the administration of extracellular vesicles derived from MSCs primed with IFN-gamma has been demonstrated to improve lung-fibrosis in preclinical studies [85,86].
Given these experiences in preclinical models, MSCs have been investigated in patients with SSc in recent decades. Published data are restricted to small case series or retrospective studies, but all reported high safety of the treatment with a very low rate of therapy-related mortality (Table 1) [57,87,88].
In a retrospective study, Liang et al. investigated the safety of a single dose of intravenous allogeneic MSCs from either a bone marrow or cord blood source; the study focused on 404 patients with autoimmune diseases treated from 2007 and 2016, including 39 cases with SSc. The primary endpoint was safety and tolerability of the treatment. The five-year overall survival was about 90%, with disease-progression or disease-related complication being the most frequent cause of death, and only one patient experiencing therapy-related death. The infection rate was up to 30%, with severe infection being around 13%, which, according to the authors, could not only be explained as a treatment complication, but might have been partially explained by disease-related immune dysfunction [90].
Zhang et al. investigated the combination of plasmapheresis and allogeneic MSC transplant in 14 patients with SSc. As per study design, patients received repeated plasmapheresis, with subsequent three cyclophosphamide pulse and a single dose of 1 × 106 cells/kg of body weight of MSC. Efficacy was measured through serologic testing and organ function analysis. The authors observed significant improvements in the mean modified Rodnan skin score after one year of follow up, as well as in pulmonary function tests in a subset of patients with lung fibrosis. Levels of anti-Scl70 autoantibodies, VEGF and TGF-beta decreased overtime [91].
More recently, Farge et al. reported the use of bone marrow-derived MSCs in 20 patients with insufficient response or contraindications to immunosuppressive therapy or HSCT. MSCs-infused doses were either 1 × 106 or 3 × 106 per body weight kilogram. The infusion was overall well tolerated, without treatment-related severe adverse events. Fifteen patients responded, with clinical benefits in terms of skin thickness [92].
Small case series have been reported as well, all showing at least transitory clinical improvements [57,88]. Optimal cell doses, administration timing, as well as source (autologous versus allogeneic) are still a matter of debate, and large, prospective clinical studies are warranted. Van Rhijn-Brouwer [93] reported the opening of a randomized, placebo-controlled study proposal (MANUS trial, NCT03211793) using intramuscular MSC injection in 20 patients suffering from SSc with DU refractory to standard treatments. Other studies have been posted on clinicaltrials.gov, aiming to investigate allogeneic MSC infusion in this setting (NCT05016804, NCT04432545, NCT04356287, NCT02213705).

4.2. Loco-Regional MSCs Use

Loco-regional MSCs use may have the advantage of treating specific SSc manifestations that severely impact quality of life, like ischaemic ulcers and skin fibrosis, in a targeted way. A promising MSCs source for local treatments appears to be the adipose tissue (AT), a tissue enriched in MSCs 500-fold greater than BM, AT-MSCs that are more easily available with painfulness collection procedures and minimal ethical considerations. AT-MSCs can be collected through liposuction from subcutaneous AT of abdominal wall or AT biopsy and expanded in vitro [83]. The harvested AT is composed of mature adipocytes (90%), extracellular matrix, and a stromal vascular fraction (SVF) which consists of AT-MSCs, along with endothelial progenitor cells, immune cells, fibroblasts, smooth muscle cells, mature endothelial cells, pericytes and cells not characterized yet [94]. SVF contains a percentage of MSCs estimated at 2–10% [95].
The regenerative potential of AT-MSCs is dependent on the paracrine effects of their secretome, constituted by a wide range of chemokines, cytokines, and protein growth factors: prostaglandin 2, vascular endothelial growth factor (VEGF) and interleukin (IL)-4, IL-6, IL-10 and IL-1 receptor antagonist [96,97]. All together, these soluble mediators sustain angiogenesis and tissue remodeling and suppress local inflammatory responses based on the local environment. Indeed, hypoxic conditions enhance a pro-angiogenic profile of AT-MSCs [98,99]. To obtain SVF, some authors used the commercially available Cytori therapeutics Celution800/CRS device (Cytori Therapeutics, San Diego, CA, USA) that processes a fat aspirate to remove fat and lipids and extract SVF or, as referred to by the company, “adipose derived regenerative cells (ADRC)”. The advantage of this technique is that removal of fat cells allows for the injection of the processed SVF or ADRC directly into arteries without risk of fat embolism. However, in order to reduce the time and costs of carrying out fat grafting without a cell lab, other authors centrifuged the harvested fat tissue and eliminated the upper oily supernatant as well as blood and debris at the bottom of the centrifuge. Only the middle layer containing adipose-derived cell fractions, such as AT-MSCs and endothelial progenitor cells, is then used for loco-regional use.
AT-MSCs have already been applied to regenerate tissues in many diseases like cardiovascular diseases, breast reconstruction after radiation and burn injuries [100,101,102]. In SSc, AT-MSCs have been used locally to treat skin fibrosis and DU, both responsible for important disability and morbidity. First studies have been conducted in localized scleroderma lesions, i.e., linear scleroderma and morphea, and showed an improvement in skin elasticity and appearance [103]. Granel et al. described a significant improvement in hand skin elasticity and function [104], later confirmed by other authors [105,106,107]. In addition, several authors reported improved labial rhyme opening through reduced perioral fibrosis and neoproliferation of dermal capillaries [108,109,110,111,112]. More recently, two controlled trials failed to reach the primary endpoint of significant improvement of hand function in SSc patients treated with SVF [113,114]. Larger trials are needed to better understand the therapeutic efficacy of AT-MSCs injection on skin fibrosis of SSc patients. Really, the micro-environment of the site of injection may have compromised the regenerative capacity of AT-MSCs or induced a myo-fibroblast-like differentiation; in addition, the use of SVF instead of whole fat may have contributed. Indeed, it is thought that SVF may be superior, since adipocytes and other cells are eliminated leaving higher concentrations of MSCs, but clear demonstration is lacking [115].
The pro-angiogenic properties of AT-MSCs have been exploited through subcutaneous injection in patients affected by DU resistant to standard treatment, with different protocols and wide variations in the harvesting, processing, and injection techniques. The use of AT-MSCs as DU therapy has been encouraged by clinical data obtained from intramuscular injection of BM-MSCs in 49 patients with DU enrolled in five different studies [116,117,118,119,120].
Bank et al. [121] were the first authors to treat with autologous fat grafting primary and secondary Raynaud’s phenomenon. In this way, the authors have observed a significant decrease in DU with minor side effects (transient numbness, cellulitis responsive at antibiotics in 1 case). Subsequently, two pilot studies demonstrated the efficacy of autologous fat grafting in inducing ulcer healing when injected at the border of the larger ulcers or at the base of the corresponding finger. Del Bene et al. treated 9 SSc patients for a total of 15 ulcers, achieving the healing of 10 DU and the size reduction above 50% of 2 DU in 8–12 weeks, with pain improvement in almost all patients. Only DUs that were located in the lower extremity or associated with atherosclerosis did not heal [122]. Del Papa et al. treated 15 SSc patients with long-lasting DU, achieving healing in all the patients in 2–7 weeks and with a significant reduction of pain in a few weeks. Interestingly, they also observed an increase in nail-fold capillaries at capillaroscopy at 3 and 6 months, and a significant after-treatment reduction of digit artery resistivity measured by high-resolution echo-color-Doppler, strongly supporting the pro-angiogenic efficacy of regional autologous fat grafting [123].
Encouraged by these data, in order to overcome pilot study limits, a monocentric randomized controlled study has been conducted. SSc patients were randomly assigned to receive either autologous fat grafting (n = 25) or a sham procedure with saline solution injection (n = 13). DU healing occurred in 23/25 and 1/13 patients treated with fat grafting and sham procedure, respectively. The 12 patients who received the unsuccessful sham procedure subsequently underwent rescue autologous fat grafting with DU healing in all of the patients within 8 weeks. The authors also confirmed significant pain improvement and an increase in nailfold capillaries already observed in the pilot study [124].
These studies have demonstrated that AT-MSCs therapies based on subcutaneous injection are safe and efficacious in treating microvascular complications of SSc such as long-lasting DU and could be the milestone for future clinical trials and applications.

5. Regulatory T Cells and Chimeric Antigen Receptor T Cells

Regulatory T cells (Tregs), a subtype of CD4 T helper cells with immune suppressive properties, are dysfunctional in several autoimmune syndromes [125]. The frequency of circulating Tregs was found to be inversely correlated with the disease activity and severity of SSc patients [126]. Theoretically, administering Tregs in this population can potentially lead to more tolerogenic micro-environment. Kamio and colleagues reported fibrosis regression after the infusion of splenic-derived Tregs in an animal model of bleomycin-induced pulmonary fibrosis [127]. Initial studies in humans using T regs (or IL-2, with subsequent boost of T regs) have focused on GvHD prophylaxis and treatment in allogeneic transplant recipients [128,129,130,131]. Despite being safe overall, polyclonal Tregs mediated sub-optimal responses in initial clinical trials, mainly due to the low amount of disease-relevant antigen-specific T cells [132,133]. A phase I/II clinical trial is currently being conducted to evaluate the tolerability and efficacy of autologous Tregs in patients with SSc (NCT05214014).
Chimeric antigen receptor (CAR) T cells are an engineered cellular product that combine B-cell antibody-based antigen recognition with T-cell cytotoxicity. This technology is redefining therapeutic strategies in the onco-hematologic field; however, the CAR ability of conferring new antigen-specificity while boosting cellular activation can be potentially employed in other setting. Indeed, in autoimmune diseases, the abrogation of autoreactive cells obtained through CAR T cells may be more profound and prolonged and might overcome the inaccessibility of inflamed tissues compared to standard drug-based approach.
In preclinical studies on models of autoimmunity, Ellebrecht et al. reported chimeric autoantibody receptors (CAARs), in which the engineered chimeric receptor displays the target protein of autoantibodies in its extracellular domain; through this mechanism, CAAR T cell would specifically bind the B-cell receptors of an autoreactive B cell, triggering its apoptosis and reducing its autoantibody production [134]. Different autoantibodies described in SSc appear to be associated with different clinical presentation and might be investigated for developing SSc-specific CAAR T cells. Whereas in some autoimmune disease specific antigen targets can be identified (e.g., insulin in type 1 diabetes), in other subtypes, including systemic sclerosis, the lack of one single autoantigen represents an obstacle for CAR-T cells’ efficacy.
CAR-T cells are also being studied as an anti-fibrotic approach. An interesting preclinical analysis published by Aghajanian and colleagues reported the use of CAR-T cells directed against fibroblast activation protein in a mouse model of cardiac fibrosis. In this model, CAR-T CD8+ cells successfully ablated cardiac fibroblast expressing xenogeneic antigen [135]. This approach has been further explored by the same group through the adoption of transient anti-fibrotic CAR-T cells in vivo by delivering modified messenger RNA in T cell–targeted lipid nanoparticles in order to improve its safety [136].
Despite these promising results in pre-clinical models, clinical trials in patients with connective tissue diseases are still preliminary. Following the positive results of the first CAR-T-cell therapy in systemic lupus erythematosus [125], the authors recently presented an update of five patients with multi-organ involvement and insufficient response to multiple previous therapies [137]. All patients received autologous CD19-directed CAR-T cells, without developing relevant toxicities, and only three patients experienced fever (cytokine-release syndrome grade 1), one received a single infusion of tocilizumab with symptoms relief; no infection occurred. After a follow-up of three months, circulating CAR-T cells were still detectable, along with a re-emergence of B cells after a median 110 of days post-infusion. All patients had a normalization of serum double-stranded DNA antibodies and complement levels and, most importantly, the resolution of nephritis and other disease-related symptoms. These data suggest a rapid response of autoimmune disease to CAR-T-cell therapy, although follow up is needed to determine long-term efficacy.
Major concerns regarding CD19-directed CAR-T cells is prolonged B-cell depletion and the subsequent increase of infection rate, which is already consistently high in population autoimmune diseases. In this regard, alternative approaches include T regs, a potential platform for CAR construct engineering with less B-cell-depleting capacity and more immune-modulating properties. CAR-T regs might potentially target either autoreactive B-cell markers (e.g., CD19), or antigens expressed on the tissue under autoimmune attack, or pathologic immune-complexes [138]. Elinav and colleagues firstly reported the use of CAR-T regs in a murine model of autoimmune colitis, showing their ability to specifically modulate pathologic effector cells, improving colitis-associated signs [139]. Other authors have investigated CAR-T regs in autoimmune models: Blat et al. reported decreased severity of ulcerative colitis after infusion of carcinoembryonic antigen-specific CAR-T regs [140], whereas Tenspolde et al. reported safety and long-term in vivo persistence of insulin-specific CAR-T regs in type 1 diabetes mellitus [141]. Compared to unmanipulated T regs, potential advantages of CAR-T reg technology include the ability to redirect T regs’ suppressive capacities while increasing the number of antigen-specific cells to be transferred to the patient. Compared to other CAR-T products, CAR T regs might have a higher safety profile in terms of off-target toxicity and effect on immune competence [138].
Ongoing studies using CAR-T cells directed against CD19 and B-cell maturation antigen (BCMA) in patients with SSc are available on clinicaltrials.gov (NCT05085444).

6. Tolerogenic Dendritic Cells

The surrounding micro-environment can skew the maturation path of immature dendritic cells toward either an immunogenic or a tolerogenic, semi-mature phenotype. Tolerogenic dendritic cells (tolDCs) play central roles in maintaining peripheral tolerance homeostasis, through secretion of anti-inflammatory cytokines (e.g., IL-10) and the expression of inhibitory molecules during antigen presentation. Indeed, after cell-to-cell interaction between T cells and tolDC, the absence of co-stimulatory molecules on tolDC prevents T-cell activation, whereas the presence of inhibitory molecules induces T-cell anergy or differentiation toward T regs.
Many protocols for in vitro and in vivo generation of tolDCs from human monocytes have been investigated, mainly in animal models. The characterization of tolDC phenotype has been reported by Comi and colleagues [142]. In this context, tolDCs loaded with disease-specific target antigens have the potential to reset and re-educate the dysfunctional immune system by abrogating pathological autoreactive T cells and promoting their anergy or differentiation toward regulatory phenotype. In a murine model of multiple sclerosis, Mansilla and colleagues reported that infusion of tolDCs loaded with myelin oligodendrocyte glycoprotein increased regulatory T cells population and decreased autoreactive T cells’ clones, which translated into neurological improvements [143]. After infusion, tolDCs have been traced to several organs, including lungs, kidney, liver, spleen, lymph nodes, thymus, bone marrow and central nervous system. However, different generation platforms as well as route of administration appear to influence tolDCs anti-inflammatory properties, their expression of trafficking chemokine receptors and their migration capacity into target organs [144,145].
TolDCs have been investigated in phase I studies in patients with multiple sclerosis, autoimmune arthritis, Crohn’s disease, and type 1 diabetes [146]; these preliminary analyses proved that tolDC administration was overall safe, determined a trend of decrease of pro-inflammatory cytokines and T cells and increase of T regs, as observed in murine models, and improved clinical symptoms. However, standardization of protocols for tolDC generation and administration is needed before their routine clinical use is possible, as well as better knowledge of immune monitoring to measure in vivo biological effect. No studies on tolDCs-based therapy for SSc have been published yet.

7. Open Questions and Future Perspectives

The biological complexity and the clinical heterogeneity of SSc raise several challenges to finding the optimal treatment (Figure 3). Autoreactive B- and T-cell clones and the dysregulation of adaptive and innate immune systems play central roles in the disease pathogenesis. Several therapeutic approaches to target them have been investigated; however, as of now, there are no biologic or immunosuppressive drugs able to effectively improve long-term drug-free survival [147].
The lack of effective therapies has guided several approaches toward the development of cell-based therapies able to restore self-tolerance [10]. Autologous hematopoietic stem cell transplant has been investigated in SSc patients in order to obtain profound immune ablation of self-reactive cells. Several phase III trials proved its benefit over standard immunosuppressive drugs in terms of long-term disease control. However, patients with severe cardiopulmonary dysfunction who undergo HSCT are at higher risk of transplant-related morbidity and mortality, thus, we could speculate that HSCT should be offered at earlier stages of their disease before the development of cardiopulmonary impairment. The appropriate patient selection and the investigation of patient-tailored, low intensity conditioning regimen represent important steps in the transplant algorithm for SSc.
Efforts have been made to investigate other non-transplant, cell-based therapies. Despite the increasing knowledge of SSc physiology, it is still unclear which patients would not have sustained response from immunosuppressants and thus could benefit from early use of cellular-based treatments. Among all, mesenchymal stem cells have raised significant interest: along with their tolerogenic properties, MSC-based treatments showed an interesting potential as regenerative therapies, able to repair tissues already damaged, without conferring the potential toxicities secondary to conditioning regimen and aplasia of HSCT. Both local and systemic use of MSCs have been studied in phase I and II studies, documenting their overall safety and potential efficacy in stabilizing, or improving, the disease. Systemic therapies may have application in diffuse cutaneous thickening and internal organ fibrosis such as interstitial lung disease, one of the most frequent causes of mortality in SSc patients and an exclusion criterion for HSCT, when extended. Local MSCs therapies have been applied in digital ulcers and skin fibrosis (mainly perioral) with improvement of both vascularization and elasticity. These techniques could be an add-on therapy to standard drug treatments in refractory disease. MSCs exert their regenerative properties also trough their secretome, that prolong their biological effect in situ. More studies are needed in order to evaluate the persistence of MSCs engrafted or infused and the long-term effects of these cells in SSc patients.
Another emerging potential approach could be the use of T-cell therapies. The use of polyclonal T regulatory cells is intriguing as, theoretically, restoration of this cellular subset which is dysfunctional in patients with SSc might provide immune modulation and disease control. Unfortunately, challenges in in vitro expansion and long-term persistence, as well as difficulties in the identification of specific antigens, have significantly slowed T-regs introduction in clinical fields. Results from ongoing studies are awaited.
Engineering of T cells through the creation of chimeric antigen receptors has led to substantial changes in the treatment of onco-hematologic diseases. CAR-T cells have the advantage of recognizing antigen in an HLA-independent fashion. Targeting B-cell-specific antigens like CD19 and B-cell maturation antigen might determine a broad depletion of autoreactive B cells, also within affected tissues, thus resetting the autoimmune process. However, these benefits might be counterbalanced by profound normal B-cell depletion, as seen in hematologic patients receiving CAR-T cells, leading to prolonged infectious risk. Recent reports of patients with systemic lupus erythematosus receiving CD19-CAR-T-cell therapy are promising, showing sustained disease remission and low toxicity [137,148]. However, more clinical studies are warranted to determine the long-term safety in patients with autoimmune diseases. Using T regs as substrate for CAR-T cells might be safer in this context, as they do not have a cytotoxic profile and could potentially restore immunologic tolerance. Choice of alternative CAR-T-cell target is also a matter of debate: fibroblasts are a potential targetable pathway, as shown by Aghajanian et al. [135] in murine models of cardiac fibrosis, although it is unclear, given the complex pathophysiology of SSc, if targeting only the final steps of the biological process could translate into sustained clinical resolution. Nevertheless, the CAR-T-cell strategy could potentially be similarly effective as autologous HSCT, yet with less toxicity, and might be a valid alternative in frail patients who cannot undergo HSCT. Also, modulation of chemotherapy dosages in the lymphodepletion regimen is being evaluated in onco-hematologic patients receiving CAR-T cells and could translate into a less toxic preparative regimen.
In conclusion, at this stage, more data are needed to draw conclusions regarding which patients might benefit from which approach. Treatment for SSc is an evolving field, and new approaches are currently under evaluation. Better understanding of the pathologic mechanisms will lead to the development of new, specific therapies to target cellular interactions and to impact clinical outcomes.

Author Contributions

R.G. and N.D.P. led on concept and design. E.X. and A.M. provided coordination and data analysis, and contributed to the analysis and the interpretation of data, writing sections of the manuscript. R.G., N.D.P. and T.A. provided expert analytical feedback and were involved in reviewing and editing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Acknowledgments

The authors contribute this article on behalf of ADWP of the EBMT. The authors thank Manuela Badoglio and Myriam Labopin in the EBMT Paris Office for provision of data from the EBMT registry, EBMT centers for their contributions to the registry and those active in the ADWP.

Conflicts of Interest

R.G. discloses speaking honoraria from Biotest, Pfizer, Medac, and Magenta. T.A. declares speaking honoraria from Amgen, Roche, and Pfizer, travel grants from Neovii, and study support from Amgen, Janssen-Cilag, and Miltenyi Biotec. A.M., E.X. and N.D.P. do not declare conflicts of interests.

Ethics Statement

This review was led and supported by the EBMT-ADWP. The EBMT provided resources via the working party, data office, and registry. Other than EBMT support, there is no funding body supporting these guidelines, commercial or otherwise.

Additional Information

Correspondence and requests for materials should be addressed to A.M. and E.X.

References

  1. Gabrielli, A.; Avvedimento, E.V.; Krieg, T. Scleroderma. N. Engl. J. Med. 2009, 360, 1989–2003. [Google Scholar] [CrossRef] [PubMed]
  2. Desbois, A.C.; Cacoub, P. Systemic sclerosis: An update in 2016. Autoimmun. Rev. 2016, 15, 417–426. [Google Scholar] [CrossRef] [Green Version]
  3. Elhai, M.; Meune, C.; Boubaya, M.; Avouac, J.; Hachulla, E.; Balbir-Gurman, A.; Riemekasten, G.; Airò, P.; Joven, B.; Vettori, S.; et al. Mapping and predicting mortality from systemic sclerosis. Ann. Rheum. Dis. 2017, 76, 1897–1905. [Google Scholar] [CrossRef] [PubMed]
  4. Poudel, D.R.; Jayakumar, D.; Danve, A.; Sehra, S.T.; Derk, C.T. Determinants of mortality in systemic sclerosis: A focused review. Rheumatol. Int. 2018, 38, 1847–1858. [Google Scholar] [CrossRef] [PubMed]
  5. Murdaca, G.; Contatore, M.; Gulli, R.; Mandich, P.; Puppo, F. Genetic factors and systemic sclerosis. Autoimmun. Rev. 2016, 15, 427–432. [Google Scholar] [CrossRef]
  6. De Martinis, M.; Ciccarelli, F.; Sirufo, M.M.; Ginaldi, L. An overview of environmental risk factors in systemic sclerosis. Expert Rev. Clin. Immunol. 2016, 12, 465–478. [Google Scholar] [CrossRef]
  7. Marie, I.; Gehanno, J.F. Environmental risk factors of systemic sclerosis. Semin. Immunopathol. 2015, 37, 463–473. [Google Scholar] [CrossRef] [PubMed]
  8. Benfaremo, D.; Svegliati, S.; Paolini, C.; Agarbati, S.; Moroncini, G. Systemic Sclerosis: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2022, 10, 163. [Google Scholar] [CrossRef] [PubMed]
  9. Denton, C.P.; Khanna, D. Systemic sclerosis. Lancet 2017, 390, 1685–1699. [Google Scholar] [CrossRef]
  10. Mosanya, C.H.; Isaacs, J.D. Tolerising cellular therapies: What is their promise for autoimmune disease? Ann. Rheum. Dis. 2019, 78, 297–310. [Google Scholar] [CrossRef]
  11. Snowden, J.A.; Sánchez-Ortega, I.; Corbacioglu, S.; Basak, G.W.; Chabannon, C.; de la Camara, R.; Dolstra, H.; Duarte, R.F.; Glass, B.; Greco, R.; et al. Indications for haematopoietic cell transplantation for haematological diseases, solid tumours and immune disorders: Current practice in Europe, 2022. Bone Marrow Transplant. 2022, 57, 1217–1239. [Google Scholar] [CrossRef]
  12. Alexander, T.; Greco, R. Hematopoietic stem cell transplantation and cellular therapies for autoimmune diseases: Overview and future considerations from the Autoimmune Diseases Working Party (ADWP) of the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 2022, 57, 1055–1062. [Google Scholar] [CrossRef] [PubMed]
  13. Sullivan, K.M.; Goldmuntz, E.A.; Furst, D.E. Autologous Stem-Cell Transplantation for Severe Scleroderma. N. Engl. J. Med. 2018, 378, 1066–1067. [Google Scholar] [CrossRef]
  14. Sullivan, K.M.; Goldmuntz, E.A.; Keyes-Elstein, L.; McSweeney, P.A.; Pinckney, A.; Welch, B.; Mayes, M.D.; Nash, R.A.; Crofford, L.J.; Eggleston, B.; et al. Myeloablative Autologous Stem-Cell Transplantation for Severe Scleroderma. N. Engl. J. Med. 2018, 378, 35–47. [Google Scholar] [CrossRef]
  15. Burt, R.K.; Shah, S.J.; Dill, K.; Grant, T.; Gheorghiade, M.; Schroeder, J.; Craig, R.; Hirano, I.; Marshall, K.; Ruderman, E.; et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): An open-label, randomised phase 2 trial. Lancet 2011, 378, 498–506. [Google Scholar] [CrossRef]
  16. van Laar, J.M.; Farge, D.; Sont, J.K.; Naraghi, K.; Marjanovic, Z.; Larghero, J.; Schuerwegh, A.J.; Marijt, E.W.; Vonk, M.C.; Schattenberg, A.V.; et al. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: A randomized clinical trial. JAMA 2014, 311, 2490–2498. [Google Scholar] [CrossRef]
  17. Burt, R.K.; Han, X.; Quigley, K.; Arnautovic, I.; Shah, S.J.; Lee, D.C.; Freed, B.H.; Jovanovic, B.; Helenowski, I.B. Cardiac safe hematopoietic stem cell transplantation for systemic sclerosis with poor cardiac function: A pilot safety study that decreases neutropenic interval to 5 days. Bone Marrow Transplant. 2021, 56, 50–59. [Google Scholar] [CrossRef] [PubMed]
  18. Alexander, T.; Greco, R.; Snowden, J.A. Hematopoietic Stem Cell Transplantation for Autoimmune Disease. Annu. Rev. Med. 2021, 72, 215–228. [Google Scholar] [CrossRef]
  19. Cencioni, M.T.; Genchi, A.; Brittain, G.; de Silva, T.I.; Sharrack, B.; Snowden, J.A.; Alexander, T.; Greco, R.; Muraro, P.A. Immune Reconstitution Following Autologous Hematopoietic Stem Cell Transplantation for Multiple Sclerosis: A Review on Behalf of the EBMT Autoimmune Diseases Working Party. Front. Immunol. 2021, 12, 813957. [Google Scholar] [CrossRef]
  20. Bohgaki, T.; Atsumi, T.; Bohgaki, M.; Furusaki, A.; Kondo, M.; Sato-Matsumura, K.C.; Abe, R.; Kataoka, H.; Horita, T.; Yasuda, S.; et al. Immunological reconstitution after autologous hematopoietic stem cell transplantation in patients with systemic sclerosis: Relationship between clinical benefits and intensity of immunosuppression. J. Rheumatol. 2009, 36, 1240–1248. [Google Scholar] [CrossRef]
  21. Arruda, L.C.M.; Malmegrim, K.C.R.; Lima-Júnior, J.R.; Clave, E.; Dias, J.B.E.; Moraes, D.A.; Douay, C.; Fournier, I.; Moins-Teisserenc, H.; Alberdi, A.J.; et al. Immune rebound associates with a favorable clinical response to autologous HSCT in systemic sclerosis patients. Blood Adv. 2018, 2, 126–141. [Google Scholar] [CrossRef] [Green Version]
  22. Roord, S.T.; de Jager, W.; Boon, L.; Wulffraat, N.; Martens, A.; Prakken, B.; van Wijk, F. Autologous bone marrow transplantation in autoimmune arthritis restores immune homeostasis through CD4+CD25+Foxp3+ regulatory T cells. Blood 2008, 111, 5233–5241. [Google Scholar] [CrossRef] [Green Version]
  23. Hendrawan, K.; Visweswaran, M.; Ma, D.D.F.; Moore, J.J. Tolerance regeneration by T regulatory cells in autologous haematopoietic stem cell transplantation for autoimmune diseases. Bone Marrow Transplant. 2020, 55, 857–866. [Google Scholar] [CrossRef]
  24. Burt, R.K.; Traynor, A.E.; Pope, R.; Schroeder, J.; Cohen, B.; Karlin, K.H.; Lobeck, L.; Goolsby, C.; Rowlings, P.; Davis, F.A.; et al. Treatment of autoimmune disease by intense immunosuppressive conditioning and autologous hematopoietic stem cell transplantation. Blood 1998, 92, 3505–3514. [Google Scholar] [CrossRef]
  25. Binks, M.; Passweg, J.R.; Furst, D.; McSweeney, P.; Sullivan, K.; Besenthal, C.; Finke, J.; Peter, H.H.; van Laar, J.; Breedveld, F.C.; et al. Phase I/II trial of autologous stem cell transplantation in systemic sclerosis: Procedure related mortality and impact on skin disease. Ann. Rheum. Dis. 2001, 60, 577–584. [Google Scholar] [CrossRef]
  26. Farge, D.; Marolleau, J.P.; Zohar, S.; Marjanovic, Z.; Cabane, J.; Mounier, N.; Hachulla, E.; Philippe, P.; Sibilia, J.; Rabian, C.; et al. Autologous bone marrow transplantation in the treatment of refractory systemic sclerosis: Early results from a French multicentre phase I-II study. Br. J. Haematol. 2002, 119, 726–739. [Google Scholar] [CrossRef]
  27. Assassi, S.; Wang, X.; Chen, G.; Goldmuntz, E.; Keyes-Elstein, L.; Ying, J.; Wallace, P.K.; Turner, J.; Zheng, W.J.; Pascual, V.; et al. Myeloablation followed by autologous stem cell transplantation normalises systemic sclerosis molecular signatures. Ann. Rheum. Dis. 2019, 78, 1371–1378. [Google Scholar] [CrossRef]
  28. Burt, R.K.; Oliveira, M.C.; Shah, S.J.; Moraes, D.A.; Simoes, B.; Gheorghiade, M.; Schroeder, J.; Ruderman, E.; Farge, D.; Chai, Z.J.; et al. Cardiac involvement and treatment-related mortality after non-myeloablative haemopoietic stem-cell transplantation with unselected autologous peripheral blood for patients with systemic sclerosis: A retrospective analysis. Lancet 2013, 381, 1116–1124. [Google Scholar] [CrossRef]
  29. Farge, D.; Burt, R.K.; Oliveira, M.C.; Mousseaux, E.; Rovira, M.; Marjanovic, Z.; de Vries-Bouwstra, J.; Del Papa, N.; Saccardi, R.; Shah, S.J.; et al. Cardiopulmonary assessment of patients with systemic sclerosis for hematopoietic stem cell transplantation: Recommendations from the European Society for Blood and Marrow Transplantation Autoimmune Diseases Working Party and collaborating partners. Bone Marrow Transplant. 2017, 52, 1495–1503. [Google Scholar] [CrossRef] [Green Version]
  30. Henes, J.; Oliveira, M.C.; Labopin, M.; Badoglio, M.; Scherer, H.U.; Del Papa, N.; Daikeler, T.; Schmalzing, M.; Schroers, R.; Martin, T.; et al. Autologous stem cell transplantation for progressive systemic sclerosis: A prospective non-interventional study from the European Society for Blood and Marrow Transplantation Autoimmune Disease Working Party. Haematologica 2021, 106, 375–383. [Google Scholar] [CrossRef] [PubMed]
  31. Ayano, M.; Tsukamoto, H.; Mitoma, H.; Kimoto, Y.; Akahoshi, M.; Arinobu, Y.; Miyamoto, T.; Horiuchi, T.; Niiro, H.; Nagafuji, K.; et al. CD34-selected versus unmanipulated autologous haematopoietic stem cell transplantation in the treatment of severe systemic sclerosis: A post hoc analysis of a phase I/II clinical trial conducted in Japan. Arthritis Res. Ther. 2019, 21, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Oliveira, M.C.; Labopin, M.; Henes, J.; Moore, J.; Del Papa, N.; Cras, A.; Sakellari, I.; Schroers, R.; Scherer, H.U.; Cuneo, A.; et al. Does ex vivo CD34+ positive selection influence outcome after autologous hematopoietic stem cell transplantation in systemic sclerosis patients? Bone Marrow Transplant. 2016, 51, 501–505. [Google Scholar] [CrossRef] [Green Version]
  33. Greco, R.; Alexander, T.; Burman, J.; Del Papa, N.; de Vries-Bouwstra, J.; Farge, D.; Henes, J.; Kazmi, M.; Kirgizov, K.; Muraro, P.A.; et al. Hematopoietic stem cell transplantation for autoimmune diseases in the time of COVID-19: EBMT guidelines and recommendations. Bone Marrow Transplant. 2021, 56, 1493–1508. [Google Scholar] [CrossRef]
  34. Shiratsuchi, M.; Motomura, S.; Abe, Y.; Shiokawa, S.; Nishimura, J. Long-term follow-up after nonmyeloablative allogeneic hematopoietic stem cell transplantation for systemic sclerosis. Clin. Rheumatol. 2008, 27, 1207–1209. [Google Scholar] [CrossRef]
  35. Khorshid, O.; Hosing, C.; Bibawi, S.; Ueno, N.; Reveille, J.; Mayes, M.D.; Champlin, R.E. Nonmyeloablative stem cell transplant in a patient with advanced systemic sclerosis and systemic lupus erythematosus. J. Rheumatol. 2004, 31, 2513–2516. [Google Scholar]
  36. Nash, R.A.; McSweeney, P.A.; Nelson, J.L.; Wener, M.; Georges, G.E.; Langston, A.A.; Shulman, H.; Sullivan, K.M.; Lee, J.; Henstorf, G.; et al. Allogeneic marrow transplantation in patients with severe systemic sclerosis: Resolution of dermal fibrosis. Arthritis Rheum. 2006, 54, 1982–1986. [Google Scholar] [CrossRef] [Green Version]
  37. Loh, Y.; Oyama, Y.; Statkute, L.; Verda, L.; Quigley, K.; Yaung, K.; Barr, W.; Jovanovic, B.; Burt, R.K. Non-myeloablative allogeneic hematopoietic stem cell transplantation for severe systemic sclerosis: Graft-versus-autoimmunity without graft-versus-host disease? Bone Marrow Transplant. 2007, 39, 435–437. [Google Scholar] [CrossRef]
  38. Greco, R.; Labopin, M.; Badoglio, M.; Veys, P.; Furtado Silva, J.M.; Abinun, M.; Gualandi, F.; Bornhauser, M.; Ciceri, F.; Saccardi, R.; et al. Allogeneic HSCT for Autoimmune Diseases: A Retrospective Study from the EBMT ADWP, IEWP, and PDWP Working Parties. Front. Immunol. 2019, 10, 1570. [Google Scholar] [CrossRef] [Green Version]
  39. Farge, D.; Loisel, S.; Lansiaux, P.; Tarte, K. Mesenchymal stromal cells for systemic sclerosis treatment. Autoimmun. Rev. 2021, 20, 102755. [Google Scholar] [CrossRef]
  40. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  41. Wang, M.; Xu, X.; Lei, X.; Tan, J.; Xie, H. Mesenchymal stem cell-based therapy for burn wound healing. Burn. Trauma 2021, 9, tkab002. [Google Scholar] [CrossRef] [PubMed]
  42. Li, A.; Guo, F.; Pan, Q.; Chen, S.; Chen, J.; Liu, H.F. Mesenchymal Stem Cell Therapy: Hope for Patients with Systemic Lupus Erythematosus. Front. Immunol. 2021, 12, 728190. [Google Scholar] [CrossRef]
  43. Lan, T.; Luo, M.; Wei, X. Mesenchymal stem/stromal cells in cancer therapy. J. Hematol. Oncol. 2021, 14, 195. [Google Scholar] [CrossRef] [PubMed]
  44. Izadi, M.; Sadr Hashemi Nejad, A.; Moazenchi, M.; Masoumi, S.; Rabbani, A.; Kompani, F.; Hedayati Asl, A.A.; Abbasi Kakroodi, F.; Jaroughi, N.; Mohseni Meybodi, M.A.; et al. Mesenchymal stem cell transplantation in newly diagnosed type-1 diabetes patients: A phase I/II randomized placebo-controlled clinical trial. Stem Cell Res. Ther. 2022, 13, 264. [Google Scholar] [CrossRef]
  45. Niess, H.; von Einem, J.C.; Thomas, M.N.; Michl, M.; Angele, M.K.; Huss, R.; Günther, C.; Nelson, P.J.; Bruns, C.J.; Heinemann, V. Treatment of advanced gastrointestinal tumors with genetically modified autologous mesenchymal stromal cells (TREAT-ME1): Study protocol of a phase I/II clinical trial. BMC Cancer 2015, 15, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Abouzaid, A.M.; El Mokadem, M.E.; Aboubakr, A.K.; Kassem, M.A.; Al Shora, A.K.; Solaiman, A. Effect of autologous fat transfer in acute burn wound management: A randomized controlled study. Burns 2022, 48, 1368–1385. [Google Scholar] [CrossRef]
  47. Ryan, J.M.; Barry, F.P.; Murphy, J.M.; Mahon, B.P. Mesenchymal stem cells avoid allogeneic rejection. J. Inflamm. 2005, 2, 8. [Google Scholar] [CrossRef] [Green Version]
  48. Caplan, H.; Olson, S.D.; Kumar, A.; George, M.; Prabhakara, K.S.; Wenzel, P.; Bedi, S.; Toledano-Furman, N.E.; Triolo, F.; Kamhieh-Milz, J.; et al. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application. Front. Immunol. 2019, 10, 1645. [Google Scholar] [CrossRef]
  49. Khan, R.S.; Newsome, P.N. A Comparison of Phenotypic and Functional Properties of Mesenchymal Stromal Cells and Multipotent Adult Progenitor Cells. Front. Immunol. 2019, 10, 1952. [Google Scholar] [CrossRef]
  50. Eliopoulos, N.; Stagg, J.; Lejeune, L.; Pommey, S.; Galipeau, J. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood 2005, 106, 4057–4065. [Google Scholar] [CrossRef]
  51. Eggenhofer, E.; Benseler, V.; Kroemer, A.; Popp, F.C.; Geissler, E.K.; Schlitt, H.J.; Baan, C.C.; Dahlke, M.H.; Hoogduijn, M.J. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front. Immunol. 2012, 3, 297. [Google Scholar] [CrossRef] [PubMed]
  52. Horwitz, E.M.; Gordon, P.L.; Koo, W.K.; Marx, J.C.; Neel, M.D.; McNall, R.Y.; Muul, L.; Hofmann, T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc. Natl. Acad. Sci. USA 2002, 99, 8932–8937. [Google Scholar] [CrossRef] [Green Version]
  53. Maria, A.T.; Toupet, K.; Maumus, M.; Fonteneau, G.; Le Quellec, A.; Jorgensen, C.; Guilpain, P.; Noël, D. Human adipose mesenchymal stem cells as potent anti-fibrosis therapy for systemic sclerosis. J. Autoimmun. 2016, 70, 31–39. [Google Scholar] [CrossRef] [PubMed]
  54. Gnecchi, M.; He, H.; Liang, O.D.; Melo, L.G.; Morello, F.; Mu, H.; Noiseux, N.; Zhang, L.; Pratt, R.E.; Ingwall, J.S.; et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat. Med. 2005, 11, 367–368. [Google Scholar] [CrossRef]
  55. Duan, X.; Lu, L.; Wang, Y.; Zhang, F.; Mao, J.; Cao, M.; Lin, B.; Zhang, X.; Shuai, X.; Shen, J. The long-term fate of mesenchymal stem cells labeled with magnetic resonance imaging-visible polymersomes in cerebral ischemia. Int. J. Nanomed. 2017, 12, 6705–6719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Guiducci, S.; Manetti, M.; Romano, E.; Mazzanti, B.; Ceccarelli, C.; Dal Pozzo, S.; Milia, A.F.; Bellando-Randone, S.; Fiori, G.; Conforti, M.L.; et al. Bone marrow-derived mesenchymal stem cells from early diffuse systemic sclerosis exhibit a paracrine machinery and stimulate angiogenesis in vitro. Ann. Rheum. Dis. 2011, 70, 2011–2021. [Google Scholar] [CrossRef]
  57. Guiducci, S.; Porta, F.; Saccardi, R.; Guidi, S.; Ibba-Manneschi, L.; Manetti, M.; Mazzanti, B.; Dal Pozzo, S.; Milia, A.F.; Bellando-Randone, S.; et al. Autologous mesenchymal stem cells foster revascularization of ischemic limbs in systemic sclerosis: A case report. Ann. Intern. Med. 2010, 153, 650–654. [Google Scholar] [CrossRef]
  58. Del Papa, N.; Quirici, N.; Soligo, D.; Scavullo, C.; Cortiana, M.; Borsotti, C.; Maglione, W.; Comina, D.P.; Vitali, C.; Fraticelli, P.; et al. Bone marrow endothelial progenitors are defective in systemic sclerosis. Arthritis Rheum. 2006, 54, 2605–2615. [Google Scholar] [CrossRef] [Green Version]
  59. Maumus, M.; Jorgensen, C.; Noël, D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: Role of secretome and exosomes. Biochimie 2013, 95, 2229–2234. [Google Scholar] [CrossRef] [Green Version]
  60. Griffin, M.; Ryan, C.M.; Pathan, O.; Abraham, D.; Denton, C.P.; Butler, P.E. Characteristics of human adipose derived stem cells in scleroderma in comparison to sex and age matched normal controls: Implications for regenerative medicine. Stem Cell Res. Ther. 2017, 8, 23. [Google Scholar] [CrossRef] [Green Version]
  61. Kastrinaki, M.C.; Sidiropoulos, P.; Roche, S.; Ringe, J.; Lehmann, S.; Kritikos, H.; Vlahava, V.M.; Delorme, B.; Eliopoulos, G.D.; Jorgensen, C.; et al. Functional, molecular and proteomic characterisation of bone marrow mesenchymal stem cells in rheumatoid arthritis. Ann. Rheum. Dis. 2008, 67, 741–749. [Google Scholar] [CrossRef] [PubMed]
  62. Larghero, J.; Farge, D.; Braccini, A.; Lecourt, S.; Scherberich, A.; Foïs, E.; Verrecchia, F.; Daikeler, T.; Gluckman, E.; Tyndall, A.; et al. Phenotypical and functional characteristics of in vitro expanded bone marrow mesenchymal stem cells from patients with systemic sclerosis. Ann. Rheum. Dis. 2008, 67, 443–449. [Google Scholar] [CrossRef]
  63. Capelli, C.; Zaccara, E.; Cipriani, P.; Di Benedetto, P.; Maglione, W.; Andracco, R.; Di Luca, G.; Pignataro, F.; Giacomelli, R.; Introna, M.; et al. Phenotypical and Functional Characteristics of In Vitro-Expanded Adipose-Derived Mesenchymal Stromal Cells from Patients with Systematic Sclerosis. Cell Transplant. 2017, 26, 841–854. [Google Scholar] [CrossRef] [Green Version]
  64. Vanneaux, V.; Farge-Bancel, D.; Lecourt, S.; Baraut, J.; Cras, A.; Jean-Louis, F.; Brun, C.; Verrecchia, F.; Larghero, J.; Michel, L. Expression of transforming growth factor β receptor II in mesenchymal stem cells from systemic sclerosis patients. BMJ Open 2013, 3, e001890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Di Benedetto, P.; Panzera, N.; Cipriani, P.; Mastroiaco, V.; Tessitore, A.; Liakouli, V.; Ruscitti, P.; Berardicurti, O.; Carubbi, F.; Guggino, G.; et al. Mesenchymal stem cells of Systemic Sclerosis patients, derived from different sources, show a profibrotic microRNA profiling. Sci. Rep. 2019, 9, 7144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Le Blanc, K.; Rasmusson, I.; Sundberg, B.; Götherström, C.; Hassan, M.; Uzunel, M.; Ringdén, O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004, 363, 1439–1441. [Google Scholar] [CrossRef]
  67. Introna, M.; Lucchini, G.; Dander, E.; Galimberti, S.; Rovelli, A.; Balduzzi, A.; Longoni, D.; Pavan, F.; Masciocchi, F.; Algarotti, A.; et al. Treatment of graft versus host disease with mesenchymal stromal cells: A phase I study on 40 adult and pediatric patients. Biol. Blood Marrow Transplant. 2014, 20, 375–381. [Google Scholar] [CrossRef] [Green Version]
  68. Muroi, K.; Miyamura, K.; Ohashi, K.; Murata, M.; Eto, T.; Kobayashi, N.; Taniguchi, S.; Imamura, M.; Ando, K.; Kato, S.; et al. Unrelated allogeneic bone marrow-derived mesenchymal stem cells for steroid-refractory acute graft-versus-host disease: A phase I/II study. Int. J. Hematol. 2013, 98, 206–213. [Google Scholar] [CrossRef]
  69. von Bahr, L.; Sundberg, B.; Lönnies, L.; Sander, B.; Karbach, H.; Hägglund, H.; Ljungman, P.; Gustafsson, B.; Karlsson, H.; Le Blanc, K.; et al. Long-term complications, immunologic effects, and role of passage for outcome in mesenchymal stromal cell therapy. Biol. Blood Marrow Transplant. 2012, 18, 557–564. [Google Scholar] [CrossRef] [Green Version]
  70. Ning, H.; Yang, F.; Jiang, M.; Hu, L.; Feng, K.; Zhang, J.; Yu, Z.; Li, B.; Xu, C.; Li, Y.; et al. The correlation between cotransplantation of mesenchymal stem cells and higher recurrence rate in hematologic malignancy patients: Outcome of a pilot clinical study. Leukemia 2008, 22, 593–599. [Google Scholar] [CrossRef] [Green Version]
  71. Kebriaei, P.; Isola, L.; Bahceci, E.; Holland, K.; Rowley, S.; McGuirk, J.; Devetten, M.; Jansen, J.; Herzig, R.; Schuster, M.; et al. Adult human mesenchymal stem cells added to corticosteroid therapy for the treatment of acute graft-versus-host disease. Biol Blood Marrow Transplant. 2009, 15, 804–811. [Google Scholar] [CrossRef] [PubMed]
  72. Kurtzberg, J.; Prockop, S.; Teira, P.; Bittencourt, H.; Lewis, V.; Chan, K.W.; Horn, B.; Yu, L.; Talano, J.A.; Nemecek, E.; et al. Allogeneic human mesenchymal stem cell therapy (remestemcel-L, Prochymal) as a rescue agent for severe refractory acute graft-versus-host disease in pediatric patients. Biol. Blood Marrow Transplant. 2014, 20, 229–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, X.; Zhang, M.; He, P. Pre-infusion single-dose mesenchymal stem cells promote platelet engraftment and decrease severe acute graft versus host disease without relapse in haploidentical peripheral blood stem cell transplantation. J. Int. Med. Res. 2020, 48, 300060520920438. [Google Scholar] [CrossRef] [PubMed]
  74. Kuzmina, L.A.; Petinati, N.A.; Parovichnikova, E.N.; Lubimova, L.S.; Gribanova, E.O.; Gaponova, T.V.; Shipounova, I.N.; Zhironkina, O.A.; Bigildeev, A.E.; Svinareva, D.A.; et al. Multipotent Mesenchymal Stromal Cells for the Prophylaxis of Acute Graft-versus-Host Disease-A Phase II Study. Stem Cells Int. 2012, 2012, 968213. [Google Scholar] [CrossRef]
  75. Maziarz, R.T.; Devos, T.; Bachier, C.R.; Goldstein, S.C.; Leis, J.F.; Devine, S.M.; Meyers, G.; Gajewski, J.L.; Maertens, J.; Deans, R.J.; et al. Single and multiple dose MultiStem (multipotent adult progenitor cell) therapy prophylaxis of acute graft-versus-host disease in myeloablative allogeneic hematopoietic cell transplantation: A phase 1 trial. Biol. Blood Marrow Transplant. 2015, 21, 720–728. [Google Scholar] [CrossRef] [Green Version]
  76. Gao, L.; Zhang, Y.; Hu, B.; Liu, J.; Kong, P.; Lou, S.; Su, Y.; Yang, T.; Li, H.; Liu, Y.; et al. Phase II Multicenter, Randomized, Double-Blind Controlled Study of Efficacy and Safety of Umbilical Cord-Derived Mesenchymal Stromal Cells in the Prophylaxis of Chronic Graft-Versus-Host Disease After HLA-Haploidentical Stem-Cell Transplantation. J. Clin. Oncol. 2016, 34, 2843–2850. [Google Scholar] [CrossRef]
  77. Lazarus, H.M.; Koc, O.N.; Devine, S.M.; Curtin, P.; Maziarz, R.T.; Holland, H.K.; Shpall, E.J.; McCarthy, P.; Atkinson, K.; Cooper, B.W.; et al. Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol. Blood Marrow Transplant. 2005, 11, 389–398. [Google Scholar] [CrossRef] [Green Version]
  78. Baron, F.; Lechanteur, C.; Willems, E.; Bruck, F.; Baudoux, E.; Seidel, L.; Vanbellinghen, J.F.; Hafraoui, K.; Lejeune, M.; Gothot, A.; et al. Cotransplantation of mesenchymal stem cells might prevent death from graft-versus-host disease (GVHD) without abrogating graft-versus-tumor effects after HLA-mismatched allogeneic transplantation following nonmyeloablative conditioning. Biol. Blood Marrow Transplant. 2010, 16, 838–847. [Google Scholar] [CrossRef] [Green Version]
  79. Zhao, F.; Zhang, Y.F.; Liu, Y.G.; Zhou, J.J.; Li, Z.K.; Wu, C.G.; Qi, H.W. Therapeutic effects of bone marrow-derived mesenchymal stem cells engraftment on bleomycin-induced lung injury in rats. Transplant. Proc. 2008, 40, 1700–1705. [Google Scholar] [CrossRef]
  80. Moodley, Y.; Atienza, D.; Manuelpillai, U.; Samuel, C.S.; Tchongue, J.; Ilancheran, S.; Boyd, R.; Trounson, A. Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am. J. Pathol. 2009, 175, 303–313. [Google Scholar] [CrossRef] [Green Version]
  81. Yang, Y.; Zhu, S.; Li, Y.; Lu, Q.; Zhang, Q.; Su, L.; Zhao, Y.; Luo, Y.; Liu, Y. Human umbilical cord mesenchymal stem cells ameliorate skin fibrosis development in a mouse model of bleomycin-induced systemic sclerosis. Exp. Ther. Med. 2020, 20, 257. [Google Scholar] [CrossRef] [PubMed]
  82. Jin, X.; Hou, J.; Zheng, K.; Wei, D.; Zhang, A.; Wang, S.; Mei, H.; Li, C.; Cheng, L.; Sun, X. Umbilical Cord Mesenchymal Stem Cells for Inhibiting the Fibrosis and Autoimmune Development in HOCl-Induced Systemic Scleroderma Mouse Model. Int. J. Stem Cells 2021, 14, 262–274. [Google Scholar] [CrossRef] [PubMed]
  83. Elessawi, D.F.; Gabr, H.; Badawy, M.M.M.; Gheita, T.A. Therapeutic potential of mesenchymal stem cells for scleroderma induced in mouse model. Tissue Cell 2021, 73, 101671. [Google Scholar] [CrossRef] [PubMed]
  84. Maria, A.T.; Toupet, K.; Bony, C.; Pirot, N.; Vozenin, M.C.; Petit, B.; Roger, P.; Batteux, F.; Le Quellec, A.; Jorgensen, C.; et al. Antifibrotic, Antioxidant, and Immunomodulatory Effects of Mesenchymal Stem Cells in HOCl-Induced Systemic Sclerosis. Arthritis Rheumatol. 2016, 68, 1013–1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Rozier, P.; Maumus, M.; Maria, A.T.J.; Toupet, K.; Jorgensen, C.; Guilpain, P.; Noël, D. Lung Fibrosis Is Improved by Extracellular Vesicles from IFNγ-Primed Mesenchymal Stromal Cells in Murine Systemic Sclerosis. Cells 2021, 10, 2727. [Google Scholar] [CrossRef] [PubMed]
  86. Huldani, H.; Abdalkareem Jasim, S.; Olegovich Bokov, D.; Abdelbasset, W.K.; Nader Shalaby, M.; Thangavelu, L.; Margiana, R.; Qasim, M.T. Application of extracellular vesicles derived from mesenchymal stem cells as potential therapeutic tools in autoimmune and rheumatic diseases. Int. Immunopharmacol. 2022, 106, 108634. [Google Scholar] [CrossRef]
  87. Christopeit, M.; Schendel, M.; Föll, J.; Müller, L.P.; Keysser, G.; Behre, G. Marked improvement of severe progressive systemic sclerosis after transplantation of mesenchymal stem cells from an allogeneic haploidentical-related donor mediated by ligation of CD137L. Leukemia 2008, 22, 1062–1064. [Google Scholar] [CrossRef]
  88. Keyszer, G.; Christopeit, M.; Fick, S.; Schendel, M.; Taute, B.M.; Behre, G.; Müller, L.P.; Schmoll, H.J. Treatment of severe progressive systemic sclerosis with transplantation of mesenchymal stromal cells from allogeneic related donors: Report of five cases. Arthritis Rheum. 2011, 63, 2540–2542. [Google Scholar] [CrossRef]
  89. Rozier, P.; Maria, A.; Goulabchand, R.; Jorgensen, C.; Guilpain, P.; Noël, D. Mesenchymal Stem Cells in Systemic Sclerosis: Allogenic or Autologous Approaches for Therapeutic Use? Front. Immunol. 2018, 9, 2938. [Google Scholar] [CrossRef] [Green Version]
  90. Liang, J.; Zhang, H.; Kong, W.; Deng, W.; Wang, D.; Feng, X.; Zhao, C.; Hua, B.; Wang, H.; Sun, L. Safety analysis in patients with autoimmune disease receiving allogeneic mesenchymal stem cells infusion: A long-term retrospective study. Stem Cell Res. Ther. 2018, 9, 312. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, H.; Liang, J.; Tang, X.; Wang, D.; Feng, X.; Wang, F.; Hua, B.; Wang, H.; Sun, L. Sustained benefit from combined plasmapheresis and allogeneic mesenchymal stem cells transplantation therapy in systemic sclerosis. Arthritis Res. Ther. 2017, 19, 165. [Google Scholar] [CrossRef] [PubMed]
  92. Farge, D.; Loisel, S.; Resche-Rigon, M.; Lansiaux, P.; Colmegna, I.; Langlais, D.; Tarte, K. Safety and preliminary efficacy of allogeneic bone marrow-derived multipotent mesenchymal stromal cells for systemic sclerosis: A single-centre, open-label, dose-escalation, proof-of-concept, phase 1/2 study. Lancet Rheumatol. 2022, 4, e91–e104. [Google Scholar] [CrossRef]
  93. van Rhijn-Brouwer, F.C.C.; Gremmels, H.; Fledderus, J.O.; Schuurman, A.H.; Bonte-Mineur, F.; Vonk, M.C.; Voskuyl, A.E.; de Vries-Bouwstra, J.K.; Coert, J.H.; Radstake, T.R.D.J.; et al. A randomised placebo-controlled double-blind trial to assess the safety of intramuscular administration of allogeneic mesenchymal stromal cells for digital ulcers in systemic sclerosis: The MANUS Trial protocol. BMJ Open 2018, 8, e020479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Gimble, J.M.; Katz, A.J.; Bunnell, B.A. Adipose-derived stem cells for regenerative medicine. Circ. Res. 2007, 100, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
  95. Bourin, P.; Bunnell, B.A.; Casteilla, L.; Dominici, M.; Katz, A.J.; March, K.L.; Redl, H.; Rubin, J.P.; Yoshimura, K.; Gimble, J.M. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: A joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy 2013, 15, 641–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Caplan, A.I.; Dennis, J.E. Mesenchymal stem cells as trophic mediators. J. Cell Biochem. 2006, 98, 1076–1084. [Google Scholar] [CrossRef] [PubMed]
  97. Kilroy, G.E.; Foster, S.J.; Wu, X.; Ruiz, J.; Sherwood, S.; Heifetz, A.; Ludlow, J.W.; Stricker, D.M.; Potiny, S.; Green, P.; et al. Cytokine profile of human adipose-derived stem cells: Expression of angiogenic, hematopoietic, and pro-inflammatory factors. J. Cell Physiol. 2007, 212, 702–709. [Google Scholar] [CrossRef]
  98. Hsiao, S.T.; Lokmic, Z.; Peshavariya, H.; Abberton, K.M.; Dusting, G.J.; Lim, S.Y.; Dilley, R.J. Hypoxic conditioning enhances the angiogenic paracrine activity of human adipose-derived stem cells. Stem Cells Dev. 2013, 22, 1614–1623. [Google Scholar] [CrossRef] [Green Version]
  99. Kondo, K.; Shintani, S.; Shibata, R.; Murakami, H.; Murakami, R.; Imaizumi, M.; Kitagawa, Y.; Murohara, T. Implantation of adipose-derived regenerative cells enhances ischemia-induced angiogenesis. Arter. Thromb. Vasc. Biol. 2009, 29, 61–66. [Google Scholar] [CrossRef] [Green Version]
  100. Perin, E.C.; Sanz-Ruiz, R.; Sánchez, P.L.; Lasso, J.; Pérez-Cano, R.; Alonso-Farto, J.C.; Pérez-David, E.; Fernández-Santos, M.E.; Serruys, P.W.; Duckers, H.J.; et al. Adipose-derived regenerative cells in patients with ischemic cardiomyopathy: The PRECISE Trial. Am. Hear. J. 2014, 168, 88–95.e82. [Google Scholar] [CrossRef]
  101. Abu-Ghname, A.; Perdanasari, A.T.; Reece, E.M. Principles and Applications of Fat Grafting in Plastic Surgery. Semin. Plast. Surg. 2019, 33, 147–154. [Google Scholar] [CrossRef] [PubMed]
  102. Chen, Y.W.; Scutaru, T.T.; Ghetu, N.; Carasevici, E.; Lupascu, C.D.; Ferariu, D.; Pieptu, D.; Coman, C.G.; Danciu, M. The Effects of Adipose-Derived Stem Cell-Differentiated Adipocytes on Skin Burn Wound Healing in Rats. J. Burn Care Res. 2017, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
  103. Scuderi, N.; Ceccarelli, S.; Onesti, M.G.; Fioramonti, P.; Guidi, C.; Romano, F.; Frati, L.; Angeloni, A.; Marchese, C. Human adipose-derived stromal cells for cell-based therapies in the treatment of systemic sclerosis. Cell Transplant. 2013, 22, 779–795. [Google Scholar] [CrossRef] [PubMed]
  104. Granel, B.; Daumas, A.; Jouve, E.; Harlé, J.R.; Nguyen, P.S.; Chabannon, C.; Colavolpe, N.; Reynier, J.C.; Truillet, R.; Mallet, S.; et al. Safety, tolerability and potential efficacy of injection of autologous adipose-derived stromal vascular fraction in the fingers of patients with systemic sclerosis: An open-label phase I trial. Ann. Rheum. Dis. 2015, 74, 2175–2182. [Google Scholar] [CrossRef] [Green Version]
  105. Guillaume-Jugnot, P.; Daumas, A.; Magalon, J.; Jouve, E.; Nguyen, P.S.; Truillet, R.; Mallet, S.; Casanova, D.; Giraudo, L.; Veran, J.; et al. Autologous adipose-derived stromal vascular fraction in patients with systemic sclerosis: 12-month follow-up. Rheumatology 2016, 55, 301–306. [Google Scholar] [CrossRef] [Green Version]
  106. Daumas, A.; Magalon, J.; Jouve, E.; Truillet, R.; Casanova, D.; Giraudo, L.; Veran, J.; Benyamine, A.; Dignat-George, F.; Magalon, G.; et al. Long-term follow-up after autologous adipose-derived stromal vascular fraction injection into fingers in systemic sclerosis patients. Curr. Res. Transl. Med. 2017, 65, 40–43. [Google Scholar] [CrossRef]
  107. Park, Y.; Lee, Y.J.; Koh, J.H.; Lee, J.; Min, H.K.; Kim, M.Y.; Kim, K.J.; Lee, S.J.; Rhie, J.W.; Kim, W.U.; et al. Clinical Efficacy and Safety of Injection of Stromal Vascular Fraction Derived from Autologous Adipose Tissues in Systemic Sclerosis Patients with Hand Disability: A Proof-Of-Concept Trial. J. Clin. Med. 2020, 9, 3023. [Google Scholar] [CrossRef]
  108. Del Papa, N.; Caviggioli, F.; Sambataro, D.; Zaccara, E.; Vinci, V.; Di Luca, G.; Parafioriti, A.; Armiraglio, E.; Maglione, W.; Polosa, R.; et al. Autologous fat grafting in the treatment of fibrotic perioral changes in patients with systemic sclerosis. Cell Transplant. 2015, 24, 63–72. [Google Scholar] [CrossRef]
  109. Onesti, M.G.; Fioramonti, P.; Carella, S.; Fino, P.; Marchese, C.; Scuderi, N. Improvement of Mouth Functional Disability in Systemic Sclerosis Patients over One Year in a Trial of Fat Transplantation versus Adipose-Derived Stromal Cells. Stem Cells Int. 2016, 2016, 2416192. [Google Scholar] [CrossRef] [Green Version]
  110. Sautereau, N.; Daumas, A.; Truillet, R.; Jouve, E.; Magalon, J.; Veran, J.; Casanova, D.; Frances, Y.; Magalon, G.; Granel, B. Efficacy of Autologous Microfat Graft on Facial Handicap in Systemic Sclerosis Patients. Plast. Reconstr. Surg. Glob. Open 2016, 4, e660. [Google Scholar] [CrossRef]
  111. Almadori, A.; Griffin, M.; Ryan, C.M.; Hunt, D.F.; Hansen, E.; Kumar, R.; Abraham, D.J.; Denton, C.P.; Butler, P.E.M. Stem cell enriched lipotransfer reverses the effects of fibrosis in systemic sclerosis. PLoS ONE 2019, 14, e0218068. [Google Scholar] [CrossRef] [PubMed]
  112. Blezien, O.; D’Andrea, F.; Nicoletti, G.F.; Ferraro, G.A. Effects of Fat Grafting Containing Stem Cells in Microstomia and Microcheilia Derived from Systemic Sclerosis. Aesthetic Plast. Surg. 2017, 41, 839–844. [Google Scholar] [CrossRef] [PubMed]
  113. Daumas, A.; Magalon, J.; Jouve, E.; Casanova, D.; Philandrianos, C.; Abellan Lopez, M.; Mallet, S.; Veran, J.; Auquit-Auckbur, I.; Farge, D.; et al. Adipose tissue-derived stromal vascular fraction for treating hands of patients with systemic sclerosis: A multicentre randomized trial Autologous AD-SVF versus placebo in systemic sclerosis. Rheumatology 2022, 61, 1936–1947. [Google Scholar] [CrossRef] [PubMed]
  114. Khanna, D.; Caldron, P.; Martin, R.W.; Kafaja, S.; Spiera, R.; Shahouri, S.; Shah, A.; Hsu, V.; Ervin, J.; Simms, R.; et al. Adipose-Derived Regenerative Cell Transplantation for the Treatment of Hand Dysfunction in Systemic Sclerosis: A Randomized Clinical Trial. Arthritis Rheumatol. 2022, 74, 1399–1408. [Google Scholar] [CrossRef]
  115. Del Papa, N.; Vitali, C.; Minniti, A.; Caporali, R. Is adipose-tissue (or its fraction) grafting really effective in the treatment of scleroderma hand? Rheumatology 2022, 61, 1756–1757. [Google Scholar] [CrossRef]
  116. Kamata, Y.; Takahashi, Y.; Iwamoto, M.; Matsui, K.; Murakami, Y.; Muroi, K.; Ikeda, U.; Shimada, K.; Yoshio, T.; Okazaki, H.; et al. Local implantation of autologous mononuclear cells from bone marrow and peripheral blood for treatment of ischaemic digits in patients with connective tissue diseases. Rheumatology 2007, 46, 882–884. [Google Scholar] [CrossRef] [Green Version]
  117. Takahashi, M.; Izawa, A.; Ishigatsubo, Y.; Fujimoto, K.; Miyamoto, M.; Horie, T.; Aizawa, Y.; Amano, J.; Minota, S.; Murohara, T.; et al. Therapeutic neovascularization by the implantation of autologous mononuclear cells in patients with connective tissue diseases. Curr. Pharm. Des. 2009, 15, 2778–2783. [Google Scholar] [CrossRef]
  118. Nevskaya, T.; Ananieva, L.; Bykovskaia, S.; Eremin, I.; Karandashov, E.; Khrennikov, J.; Mach, E.; Zaprjagaeva, M.; Guseva, N.; Nassonov, E. Autologous progenitor cell implantation as a novel therapeutic intervention for ischaemic digits in systemic sclerosis. Rheumatology 2009, 48, 61–64. [Google Scholar] [CrossRef]
  119. Ishigatsubo, Y.; Ihata, A.; Kobayashi, H.; Hama, M.; Kirino, Y.; Ueda, A.; Takeno, M.; Shirai, A.; Ohno, S. Therapeutic angiogenesis in patients with systemic sclerosis by autologous transplantation of bone-marrow-derived cells. Mod. Rheumatol. 2010, 20, 263–272. [Google Scholar] [CrossRef]
  120. Takagi, G.; Miyamoto, M.; Tara, S.; Kirinoki-Ichikawa, S.; Kubota, Y.; Hada, T.; Takagi, I.; Mizuno, K. Therapeutic vascular angiogenesis for intractable macroangiopathy-related digital ulcer in patients with systemic sclerosis: A pilot study. Rheumatology 2014, 53, 854–859. [Google Scholar] [CrossRef] [Green Version]
  121. Bank, J.; Fuller, S.M.; Henry, G.I.; Zachary, L.S. Fat grafting to the hand in patients with Raynaud phenomenon: A novel therapeutic modality. Plast. Reconstr. Surg. 2014, 133, 1109–1118. [Google Scholar] [CrossRef] [PubMed]
  122. Del Bene, M.; Pozzi, M.R.; Rovati, L.; Mazzola, I.; Erba, G.; Bonomi, S. Autologous fat grafting for scleroderma induced digital ulcers. An effective technique in patients with systemic sclerosis. Handchir. Mikrochir. Plast. Chir. 2014, 46, 242–247. [Google Scholar]
  123. Del Papa, N.; Di Luca, G.; Sambataro, D.; Zaccara, E.; Maglione, W.; Gabrielli, A.; Fraticelli, P.; Moroncini, G.; Beretta, L.; Santaniello, A.; et al. Regional implantation of autologous adipose tissue-derived cells induces a prompt healing of long-lasting indolent digital ulcers in patients with systemic sclerosis. Cell Transplant. 2015, 24, 2297–2305. [Google Scholar] [CrossRef] [PubMed]
  124. Del Papa, N.; Di Luca, G.; Andracco, R.; Zaccara, E.; Maglione, W.; Pignataro, F.; Minniti, A.; Vitali, C. Regional grafting of autologous adipose tissue is effective in inducing prompt healing of indolent digital ulcers in patients with systemic sclerosis: Results of a monocentric randomized controlled study. Arthritis Res. Ther. 2019, 21, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Ugor, E.; Simon, D.; Almanzar, G.; Pap, R.; Najbauer, J.; Németh, P.; Balogh, P.; Prelog, M.; Czirják, L.; Berki, T. Increased proportions of functionally impaired regulatory T cell subsets in systemic sclerosis. Clin. Immunol. 2017, 184, 54–62. [Google Scholar] [CrossRef]
  126. Slobodin, G.; Ahmad, M.S.; Rosner, I.; Peri, R.; Rozenbaum, M.; Kessel, A.; Toubi, E.; Odeh, M. Regulatory T cells (CD4(+)CD25(bright)FoxP3(+)) expansion in systemic sclerosis correlates with disease activity and severity. Cell. Immunol. 2010, 261, 77–80. [Google Scholar] [CrossRef]
  127. Kamio, K.; Azuma, A.; Matsuda, K.; Usuki, J.; Inomata, M.; Morinaga, A.; Kashiwada, T.; Nishijima, N.; Itakura, S.; Kokuho, N.; et al. Resolution of bleomycin-induced murine pulmonary fibrosis via a splenic lymphocyte subpopulation. Respir. Res. 2018, 19, 71. [Google Scholar] [CrossRef] [Green Version]
  128. Trzonkowski, P.; Bieniaszewska, M.; Juścińska, J.; Dobyszuk, A.; Krzystyniak, A.; Marek, N.; Myśliwska, J.; Hellmann, A. First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127- T regulatory cells. Clin. Immunol. 2009, 133, 22–26. [Google Scholar] [CrossRef]
  129. Brunstein, C.G.; Miller, J.S.; Cao, Q.; McKenna, D.H.; Hippen, K.L.; Curtsinger, J.; Defor, T.; Levine, B.L.; June, C.H.; Rubinstein, P.; et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: Safety profile and detection kinetics. Blood 2011, 117, 1061–1070. [Google Scholar] [CrossRef]
  130. Di Ianni, M.; Falzetti, F.; Carotti, A.; Terenzi, A.; Castellino, F.; Bonifacio, E.; Del Papa, B.; Zei, T.; Ostini, R.I.; Cecchini, D.; et al. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood 2011, 117, 3921–3928. [Google Scholar] [CrossRef] [Green Version]
  131. Matsuoka, K.; Koreth, J.; Kim, H.T.; Bascug, G.; McDonough, S.; Kawano, Y.; Murase, K.; Cutler, C.; Ho, V.T.; Alyea, E.P.; et al. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci. Transl. Med. 2013, 5, 179ra143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bluestone, J.A.; Buckner, J.H.; Fitch, M.; Gitelman, S.E.; Gupta, S.; Hellerstein, M.K.; Herold, K.C.; Lares, A.; Lee, M.R.; Li, K.; et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci. Transl. Med. 2015, 7, 315ra189. [Google Scholar] [CrossRef] [PubMed]
  133. Chwojnicki, K.; Iwaszkiewicz-Grześ, D.; Jankowska, A.; Zieliński, M.; Łowiec, P.; Gliwiński, M.; Grzywińska, M.; Kowalczyk, K.; Konarzewska, A.; Glasner, P.; et al. Administration of CD4. BioDrugs 2021, 35, 47–60. [Google Scholar] [CrossRef]
  134. Ellebrecht, C.T.; Bhoj, V.G.; Nace, A.; Choi, E.J.; Mao, X.; Cho, M.J.; Di Zenzo, G.; Lanzavecchia, A.; Seykora, J.T.; Cotsarelis, G.; et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science 2016, 353, 179–184. [Google Scholar] [CrossRef] [Green Version]
  135. Aghajanian, H.; Kimura, T.; Rurik, J.G.; Hancock, A.S.; Leibowitz, M.S.; Li, L.; Scholler, J.; Monslow, J.; Lo, A.; Han, W.; et al. Targeting cardiac fibrosis with engineered T cells. Nature 2019, 573, 430–433. [Google Scholar] [CrossRef] [PubMed]
  136. Rurik, J.G.; Tombácz, I.; Yadegari, A.; Méndez Fernández, P.O.; Shewale, S.V.; Li, L.; Kimura, T.; Soliman, O.Y.; Papp, T.E.; Tam, Y.K.; et al. CAR T cells produced in vivo to treat cardiac injury. Science 2022, 375, 91–96. [Google Scholar] [CrossRef] [PubMed]
  137. Mackensen, A.; Müller, F.; Mougiakakos, D.; Böltz, S.; Wilhelm, A.; Aigner, M.; Völkl, S.; Simon, D.; Kleyer, A.; Munoz, L.; et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat. Med. 2022, 28, 2124–2132. [Google Scholar] [CrossRef]
  138. Sadeqi Nezhad, M.; Seifalian, A.; Bagheri, N.; Yaghoubi, S.; Karimi, M.H.; Adbollahpour-Alitappeh, M. Chimeric Antigen Receptor Based Therapy as a Potential Approach in Autoimmune Diseases: How Close Are We to the Treatment? Front. Immunol. 2020, 11, 603237. [Google Scholar] [CrossRef]
  139. Elinav, E.; Waks, T.; Eshhar, Z. Redirection of regulatory T cells with predetermined specificity for the treatment of experimental colitis in mice. Gastroenterology 2008, 134, 2014–2024. [Google Scholar] [CrossRef] [Green Version]
  140. Blat, D.; Zigmond, E.; Alteber, Z.; Waks, T.; Eshhar, Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol. Ther. 2014, 22, 1018–1028. [Google Scholar] [CrossRef] [Green Version]
  141. Tenspolde, M.; Zimmermann, K.; Weber, L.C.; Hapke, M.; Lieber, M.; Dywicki, J.; Frenzel, A.; Hust, M.; Galla, M.; Buitrago-Molina, L.E.; et al. Regulatory T cells engineered with a novel insulin-specific chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes. J. Autoimmun. 2019, 103, 102289. [Google Scholar] [CrossRef] [PubMed]
  142. Comi, M.; Avancini, D.; Santoni de Sio, F.; Villa, M.; Uyeda, M.J.; Floris, M.; Tomasoni, D.; Bulfone, A.; Roncarolo, M.G.; Gregori, S. Coexpression of CD163 and CD141 identifies human circulating IL-10-producing dendritic cells (DC-10). Cell. Mol. Immunol. 2020, 17, 95–107. [Google Scholar] [CrossRef] [PubMed]
  143. Mansilla, M.J.; Sellès-Moreno, C.; Fàbregas-Puig, S.; Amoedo, J.; Navarro-Barriuso, J.; Teniente-Serra, A.; Grau-López, L.; Ramo-Tello, C.; Martínez-Cáceres, E.M. Beneficial effect of tolerogenic dendritic cells pulsed with MOG autoantigen in experimental autoimmune encephalomyelitis. CNS Neurosci. Ther. 2015, 21, 222–230. [Google Scholar] [CrossRef] [PubMed]
  144. Tal, O.; Lim, H.Y.; Gurevich, I.; Milo, I.; Shipony, Z.; Ng, L.G.; Angeli, V.; Shakhar, G. DC mobilization from the skin requires docking to immobilized CCL21 on lymphatic endothelium and intralymphatic crawling. J. Exp. Med. 2011, 208, 2141–2153. [Google Scholar] [CrossRef] [Green Version]
  145. Ridolfi, R.; Riccobon, A.; Galassi, R.; Giorgetti, G.; Petrini, M.; Fiammenghi, L.; Stefanelli, M.; Ridolfi, L.; Moretti, A.; Migliori, G.; et al. Evaluation of in vivo labelled dendritic cell migration in cancer patients. J. Transl. Med. 2004, 2, 27. [Google Scholar] [CrossRef] [Green Version]
  146. Morante-Palacios, O.; Fondelli, F.; Ballestar, E.; Martínez-Cáceres, E.M. Tolerogenic Dendritic Cells in Autoimmunity and Inflammatory Diseases. Trends Immunol. 2021, 42, 59–75. [Google Scholar] [CrossRef]
  147. Kowal-Bielecka, O.; Fransen, J.; Avouac, J.; Becker, M.; Kulak, A.; Allanore, Y.; Distler, O.; Clements, P.; Cutolo, M.; Czirjak, L.; et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann. Rheum. Dis. 2017, 76, 1327–1339. [Google Scholar] [CrossRef] [Green Version]
  148. Doglio, M.; Alexander, T.; Del Papa, N.; Snowden, J.A.; Greco, R. New insights in systemic lupus erythematosus: From regulatory T cells to CAR-T-cell strategies. J. Allergy Clin. Immunol. 2022, S0091-6749(22)01056-9. [Google Scholar] [CrossRef]
Figure 1. Number of autologous transplant procedures for the treatment of systemic sclerosis. Data as reported to the European Blood and Marrow Transplantation Society (EBMT).
Figure 1. Number of autologous transplant procedures for the treatment of systemic sclerosis. Data as reported to the European Blood and Marrow Transplantation Society (EBMT).
Cells 11 03346 g001
Figure 2. Cellular therapies might be adopted for both systemic and loco-regional use in patients with SSc. On the left side: systemic treatments including hematopoietic stem cell transplantation, mesenchymal stem cells, CAR-T cells and tolerogenic dendritic cells. On the right side: local treatments rely on mesenchymal stem cells obtained from adipose tissue or bone marrow successively engrafted to digits or perioral area.
Figure 2. Cellular therapies might be adopted for both systemic and loco-regional use in patients with SSc. On the left side: systemic treatments including hematopoietic stem cell transplantation, mesenchymal stem cells, CAR-T cells and tolerogenic dendritic cells. On the right side: local treatments rely on mesenchymal stem cells obtained from adipose tissue or bone marrow successively engrafted to digits or perioral area.
Cells 11 03346 g002
Figure 3. Advantages and limitations of different types of available or potential treatment options for SSc.
Figure 3. Advantages and limitations of different types of available or potential treatment options for SSc.
Cells 11 03346 g003
Table 1. Clinical trials investigating mesenchymal cells for the treatment of systemic sclerosis.
Table 1. Clinical trials investigating mesenchymal cells for the treatment of systemic sclerosis.
MSC IsolationAdministration RouteSourceTreated PatientsClinical TargetOutcomeMajor Adverse EventsReferences
BM-derived MSCsIntravenousAllo1Severe diffuse SScUlcer healing and improved skin elasticity/vascularizationNoneChristopeit et al., 2008
BM-derived MSCsIntravenousAllo5Severe diffuse SScAll patients had temporary clinical improvementMinor respiratory infectionKeyzer et al., 2011
BM-derived MSCsIntravenousAllo20Severe diffuse SSc15 patients showed sustained improvement in skin thicknessNoneFarge et al., 2022
BM-derived MSCs and peripheral blood MSCsIntramuscular injectionAuto6
(4 SSc, 2 mixed connective tissue disease)
Severe ischaemia and necrosis in fingers and/or toesPain relief in 5/6 patientsNoneKamata et al., 2007
BM-derived MSCs and peripheral blood MSCsIntramuscular injectionAuto2Severe digital and malleolar ulcersAll had improvements with ulcers healing, pain relief, reduction of RPNoneNevskaya et al., 2009
BM-derived MSCs and peripheral blood MSCsIntramuscular injectionAuto46
(24 SSc, 22 other connective tissue diseases)
Severe digital ulcers20/23 SSc patients had improvement in pain and ulcersNoneTakahashi et al., 2009
BM-derived MSCsIntramuscular injectionAuto8Severe digital ulcersAll had ulcers size and pain improvementNoneIshigatsubo et al., 2010
BM-derived MSCsIntramuscular injectionAuto40
(11 SSc, 29 with arteriosclerosis obliterans)
Severe digital ulcersAll had pain and trans-cutaneous oxygen tension improvementMajor limb amputation due to pre-existing osteomyelitisTakagi et al., 2014
Adipose derived cell fractionsSubcutaneous injectionAuto13Raynaud’s phenomenon10 patients had clinical benefit, 3 reported no changes.NoneBank et al., 2014
Adipose derived cell fractionsSubcutaneous injectionAuto20Peri-oral fibrosisAll patients had improved skin elasticity and vascularizationSmall areas of ecchymosisDel Papa et al., 2015
Adipose derived cell fractionsSubcutaneous injectionAuto15Severe digital ulcersAll patients displayed clinical benefit with fast healing of digital ulcersNoneDel Papa et al., 2015
Adipose derived cell fractions plus platelet-rich plasmaSubcutaneous injectionAuto6Peri-oral fibrosisAll patients had improved skin elasticity and vascularizationNoneVirzì et al., 2017
Adipose derived cell fractionsSubcutaneaous/perioral injectionAuto6Skin sclerodermaImprovement in 4 patients, stabilization in allNoneScuderi et al., 2013
Adipose derived cell fractionsSubcutaneous injectionAuto5Peri-oral fibrosisAll patients had improvement to perioral fibrosisNoneOnesti et al., 2016
Adipose derived cell fractions plus platelet-rich plasmaSubcutaneous injectionAuto7Peri-oral fibrosisAll patients had improvement to perioral fibrosisNoneBlezien et al., 2017
Adipose derived cell fractionsSubcutaneous injectionAuto62Peri-oral fibrosisImprovement in mouth openingSuperficial wound infectionAlmadori et al., 2019
SVFSubcutaneous injectionAuto12Severe hand dysfunctionImprovement of pain, grasping capacity, finger oedema, Raynaud’s phenomenom, quality of lifeNoneGuillaume-Jugnot et al., 2016,
Daumas et al., 2017,
Granel et al., 2015
SVFSubcutaneous injectionAuto18Severe hand dysfunctionImprovement of skin fibrosis, hand oedema, and quality of lifeNonePark et al., 2020
Adipose derived cell fractionsSubcutaneous injectionAuto9Severe digital ulcersAll patients had pain improvement, digital ulcers improvement or healingNoneDel Bene et al., 2014
Adipose derived cell fractions vs placeboSubcutaneous injectionAuto25 vs. 13Severe digital ulcers23/25 and 1/13 patients had digital ulcers improvement and healing, pain reduction and improvement on nail fold capillaroscopyNoneDel Papa et al., 2019
SVF
vs placebo
Subcutaneous injectionAuto20 vs. 20Severe hand dysfunctionImprovement of hand function in both groups, with no superiority of the SVFHypoxaemia during the surgical processDaumas et al., 2022
Adipose derived cell fractions vs placeboSubcutaneous injectionAuto48 vs. 40Severe hand dysfunctionNo improvement of hand functionAspiration pneumonia, hypotension, anginaKhanna et al., 2022
BM, bone marrow; MSCs, mesenchymal stem cells; SVF, stromal vascular fraction; SSc, systemic sclerosis. Adapted from Rozier et al. [89].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xue, E.; Minniti, A.; Alexander, T.; Del Papa, N.; Greco, R.; on behalf of The Autoimmune Diseases Working Party of the European Society for Blood and Marrow Transplantation. Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches. Cells 2022, 11, 3346. https://doi.org/10.3390/cells11213346

AMA Style

Xue E, Minniti A, Alexander T, Del Papa N, Greco R, on behalf of The Autoimmune Diseases Working Party of the European Society for Blood and Marrow Transplantation. Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches. Cells. 2022; 11(21):3346. https://doi.org/10.3390/cells11213346

Chicago/Turabian Style

Xue, Elisabetta, Antonina Minniti, Tobias Alexander, Nicoletta Del Papa, Raffaella Greco, and on behalf of The Autoimmune Diseases Working Party (ADWP) of the European Society for Blood and Marrow Transplantation (EBMT). 2022. "Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches" Cells 11, no. 21: 3346. https://doi.org/10.3390/cells11213346

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop