Rehab Y Al-Ansari01, Khalid A Al-Anazi2*
1- Consultant Hematologist, Section of Adult Hematology, Department of Internal Medicine, King Fahad Military Medical Complex, Dhahran, Saudi Arabia
2- Consultant Hemato-Oncologist and Chairman, Department of Hematology and Hematopoietic, Stem Cell Transplantation, Oncology Center, King Fahad Specialist Hospital, Saudi Arabia
*Corresponding Author: Khalid Ahmed Al-Anazi, Consultant Hemato-Oncologist and Chairman, Department of Hematology and Hematopoietic Stem Cell Transplantation, Oncology Center, King Fahad Specialist Hospital, P.O. Box: 15215, Dammam 31444, Saudi Arabia; Email: [email protected]
Published Date: 18-09-2020
Copyright© 2020 by Al-Anazi KA, et al. All rights reserved. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Abstract
Systemic sclerosis systemic is a systemic autoimmune disease that ultimately leads to severe damage and failure of multiple body organs. The disease is associated with significant morbidity and mortality as well as poor response to the available immunosuppressive therapies.
Stem cells particularly hematopoietic and mesenchymal stem cells have recently been shown to be not only safe but also efficacious in the treatment of patients with systemic sclerosis. Autologous transplantation of hematopoietic stem cells has become the standard of care for patients with severe systemic sclerosis. Mesenchymal stem cells, alone or in combination with other therapies, have been shown to be effective in the treatment of certain forms and complications of systemic sclerosis such as chronic sclerodermatous graft versus host disease. The review will discuss the various aspects of systemic sclerosis and it will highlight the role of hematopoietic as well as mesenchymal stem cells in the treatment of systemic sclerosis.
Keywords
Systemic Sclerosis; Stem Cell Therapies; Hematopoietic Stem Cells; Mesenchymal Stem Cells; Autologous Transplantation
Introduction to Systemic Sclerosis
Scleroderma or Systemic Sclerosis (SSc) is a chronic systemic autoimmune disease characterized by widespread obliterative microvasculopathy, activation of the immune system with production of autoantibodies, and variable degrees of inflammation and tissue fibrosis that ultimately lead to severe damage and failure of multiple body organs including: skin, blood vessels, lungs, heart, kidneys, Gastrointestinal Tract (GIT), and musculoskeletal system [1-4].
Scleroderma patients have defects in the number and function of circulating endothelial cells. Bone Marrow (BM)-circulating endothelial progenitor cells play a key role in blood vessel repair and neovascularization. Therefore, manipulating the production, function, and differentiation of circulating progenitor cells may represent a new therapeutic modality for treating scleroderma [5]. The Chitinase-like protein (Chi 3L1) is associated with enhanced inflammatory and fibrotic processes and the presence of progenitor cells expressing Chi 3L1 in the skin of patients with SSc appears to play a role in the initiation of the disease process [6].
Etiology and Associations of SSc
The etiology of SSC is multifactorial. The various factors that are involved in the etiology and associations of SSc are shown in Table 1 [7-16]. However, there are familial aggregates of SSc [13]. Polymorphisms of the major histocompatibility complex Human Leukocyte Antigen (HLA) particularly class II HLA have been associated with disease development [7,12,13]. Specific genes are involved particularly: STAT4, BANK1, IRF5, IRF8, TNFSF4, Interleukin (IL)-23R, IL-12RB1, IL-12RB2, ATG5, fibrillin1, MIF173, PTPN22, and BLK genes [7,9,10].
In a systematic review and a meta-analysis that included 34 published studies, the following results were obtained [8]:
- The confirmed risk factors included: female gender, age between 45 and 64 years, positive family history, and exposure to silica
- Microchimerism and exposure to organic solvents were controversial associations
There was negative correlation with alcohol consumption, cigarette smoking and infectious agents
1 | Unknown etiology |
2 | Autoimmune disorder associated with connective tissue diseases |
3 | Genetic susceptibility |
4 | Environmental exposure: vinylchloride, and organic solvents |
5 | Silicone breast implants |
6 | Infectious agents: |
a | Cytomegalovirus |
b | Epstein Barr virus |
c | Parvovirus B-19 |
d | Lyme disease |
7 | Medications: bleomycin, cocaine and pentazocine |
8 | Trauma |
9 | Radiation |
10 | Chronic graft versus host disease following allogeneic hematopoietic stem cell transplantation |
Table 1: Etiology and associations of systemic sclerosis.
Subtypes of SSc
SSc has the following subsets or subtypes [17-20]:
- Limited cutaneous SSc
- Diffuse cutaneous SSc
- Intermediate cutaneous SSc
- SSc Sine Scleroderma
Although CREST syndrome (Calcinosis, Raynaud’s phenomenon, Esophageal hypomotility, Sclerodactyly, Telangiectasia) is most confined to a single subset of SSc, it is classified as a variant of limited cutaneous SSC and may be considered as intermediate cutaneous SSc [18,20]. Localized forms of scleroderma include linear scleroderma and morphea [18]. Raynaud’s phenomenon and scleroderma, hardening of the skin, are the clinical hallmarks of SSc [18].
Chronic Sclerodermatous Graft Versus Host Disease (GVHD)
Sclerodermatous chronic GVHD is a form of chronic GVHD that is characterized by involvement of skin and subcutaneous tissues or fascia without significant visceral involvement [15]. Manifestations of sclerodermatous chronic GVHD include: (1) marked cutaneous sclerosis with skin induration or atrophy, and (2) Sjogren’s syndrome with xerostomia and xerophthalmia [16,21,22]. However, sclerodermatous chronic GVHD is believed to be an autoimmune disease or process [15]. Similarities between chronic GVHD and scleroderma include: chronic fibrosis including skin fibrosis; immunological abnormalities including autoantibodies, and involvement of mast cells [23,24]. Differences between chronic GVHD and scleroderma include: the type of collagen laid down and its precise location; distribution of inner organ involvement; absence of Raynaud’s phenomenon in chronic GVHD; lack of ischemic changes in digital extremities in chronic GVHD; and the superficial dermal microvessels have no capillary refractoriness and no loss of endothelial cell specific markers in chronic GVHD [23,24].
The use of DNA microarray technology in murine models of sclerodermatous chronic GVHD has shown that histological development of cutaneous scleroderma is accompanied by upregulated expression of many chemokines and their receptors [25]. In patients with sclerodermatous chronic GVHD, antibodies against platelet derived growth factor receptor have been reported [26]. Sclerodermatous chronic GVHD is usually refractory to steroid therapy but may benefit from tyrosine kinase inhibitors such as imatinib [26].
The factors that are associated with high incidence of sclerosis include: mobilized blood cell grafts; conditioning therapy with total body irradiation; and allografts obtained from HLA-identical sibling donors, while low incidence of sclerosis is associated with: use of HLA-mismatched donors; and use of major ABO-mismatched donors [16,27]. Patients with chronic GVHD have high incidence of sclerosis, which can cause disability but does not affect mortality or recurrence of malignancy in patients with chronic GVHD [27]. Sclerosis occurs in up to 20% of patients by 3 years after starting systemic therapy for chronic GVHD. Fibrotic manifestations of chronic GVHD include: Bronchiolitis Obliterans (BO) as well as skin thickening and fibrosis [14].
Studies in murine models of sclerodermatous GVHD have shown that:
- The main pathological changes in the skin and lungs are: monocyte infiltration, upregulation of cutaneous collagen messenger RNA
- Sclerodermatous GVHD can be prevented by the administration of TGF-β1 neutralizing antibodies which can effectively block the influx of monocyte/macrophage and T-cells into the skin and prevent new collagen synthesis by abrogation of the upregulation of TGF-β1 [28,29]. Fibrosis in sclerodermatous GVHD is driven by production of TGF-β1 by mononuclear cells [29]
Clinical Manifestations and Complications of SSc
The most typical symptoms of SSc emerge from dermal and vascular lesions [30]. Pulmonary fibrosis and pulmonary artery hypertension are the leading causes of death in patients with SSc as pulmonary complications of SSc are one of the most challenging complications of SSc [18,31]. GIT complications are the most frequent internal complications of SSc [32]. The clinical manifestations and the systemic complications of SSc are included in Table 2 [2,18,31-37]. The differential diagnosis of SSC is shown in Table 3 [38-41].
1
| Cardiovascular
| · Heart block, arrhythmias, and other conduction disturbances |
· Diastolic dysfunction, left ventricular and congestive cardiac failure | ||
· Myocarditis, cardiac fibrosis and valvular abnormalities | ||
· Pericardial effusions, vasospasm and ischemic heart disease | ||
· Raynaud’s phenomenon, digital ischemia and digital ulcers | ||
2
| Gastrointestinal
| · Gastroesophageal reflux disease with dysphagia and regurgitation |
· Gastric antral vascular ectasia | ||
· Barrett esophagus, esophageal strictures and carcinoma of esophagus | ||
· Small intestinal bacterial overgrowth and intestinal malabsorption | ||
· Chronic large intestinal pseudo-obstruction | ||
· Xerostomia, gastroparesis and delayed gastric emptying | ||
· Fecal incontinence due to loss of internal anal sphincter function | ||
3 | Genito-urinary | · Sexual dysfunction |
4
| Musculoskeletal
| · Muscle weakness and atrophy, myopathy and myositis |
· Puffy hands and fingers and shortening of distal phalanges | ||
· Flexion contractures and restriction of joint movement | ||
· Tendon friction rubs and tenosynovitis | ||
· Arthritis and erosive arthropathy | ||
5
| Pulmonary
| · Pulmonary fibrosis, interstitial pneumonitis and interstitial lung disease |
· Pulmonary arterial hypertension and restrictive ventilator defects | ||
· Pulmonary infiltrates and ground-glass opacities on chest X-ray | ||
6 | Renal | · Renal crisis, proteinuria and azotemia |
7
| Skin
| · Calcinosis, shortening of distal phalanges. telangiectasia, pruritis and thick skin tags |
· Hypopigmentation and hyperpigmentation | ||
· Tight skin leading to: mask-line face and narrowing of mouth aperture | ||
8
| Central Nervous System | · Transient ischemic attack and stroke |
· Headache, dizziness, aphasia and visual disturbances | ||
· Anxiety, depression, convulsions and coma |
Table 2: Systemic complications of systemic sclerosis.
S. No. | Differential Diagnosis |
1 | Lichen sclerosis |
2 | Morphea form of basal cell carcinoma |
3 | Post-radiation morphea |
4 | Vaccination-associated morphea |
5 | Injection of silica, paraffin and vitamin-K |
6 | Esinophilic fascitis |
7 | Porphyria |
8 | Amyloidosis |
9 | Lipodermatosclerosis |
10 | Diabetic cheiroarthropathy/cheiroarthritis |
11 | Scleromyxedema |
12 | Nephrogenic systemic fibrosis |
13 | Mucinosis associated with thyroid disorders |
14 | Scleroderma adultorum Buschke |
Table 3: Differential diagnosis of systemic sclerosis.
Treatment of SSc
SSc is a disease which is associated with significant morbidity and mortality with relatively small benefits from immunosuppressive therapies [42]. In SSc, it is difficult to develop effective disease-modifying agents and to perform randomized controlled clinical trials due to the following reasons: the clinical heterogeneity of the disease; the scarcity of objective tools to evaluate SSc; the extent of organ involvement; and the rapid progression of the disease [43,44].
So far, there is no curative or standard effective therapy for SSc [1,3]. Treatment of SSc is aimed at: improving the Quality of Life (QoL), minimizing specific organ involvement, and prevention of life-threatening complications [3]. However, the management of SSc is multidisciplinary and requires a holistic approach [2]. The available therapeutic options for SSc are shown in Table 4 [1,2,4].
1
| Treatment of Symptoms and Complications
| · Antimicrobials for infections |
· Proton pump inhibitors for gastrointestinal complications | ||
· Angiotensin-converting enzyme inhibitors and calcium channel blockers for hypertension | ||
2 | Immunosuppressive Therapies | · Cyclophosphamide, azathioprine, methotrexate and mycophenolate mofotil |
3
| Biological Therapies
| · Intravenous immunoglobulins and abatacept |
· Monoclonal antibodies such as: rituximab, tocilizumab, belimumab, inebilizumab and fresolimumab | ||
· Systemic targeted therapies including | ||
· Rapamycin | ||
· Tyrosine kinase inhibitors: imatinib, dasatinib, nilotinib | ||
4 | Stem Cell and Organ Transplantation | · Autologous hematopoietic stem cell transplantation for early diffuse systemic sclerosis |
Table 4: Available therapeutic modalities for systemic sclerosis.
Stem Cell and Their Role in the Therapy of SSc
Stem cells are a subset of biological cells in the human body that are capable of self-renewal, tissue repair, differentiation, and division into different cell lineages [45-47]. Based on their origin and potency, stem cells are classified into either embryonic and adult stem cells or unipotent, oligopotent, totipotent, multipotent, and pluripotent stem cells [45,46,48,49]. Mesenchymal Stem Cells (MSCs) are heterogenous adult multipotent stromal cells that can be isolated from various sources including: BM, Peripheral Blood (PB), Umbilical Cord Blood (UCB), and Adipose Tissue (AT) [50,51]. They have certain immunomodulatory, immunosuppressive, and antimicrobial properties that enable them to have several clinical applications including: their role in transplantation of Hematopoietic Stem Cells (HSCs) as well as regenerative medicine and treatment of autoimmune disorders [50,51].
HSCs which arise during embryogenesis sustain the production of all blood cells in humans throughout their lifetime [52-56]. They are the only cells within the hematopoietic system that possess the ability of multipotency and self-renewal [54-57]. The local BM microenvironment or the niche provides HSCs with the crucial and indispensable factors that are needed for self-renewal and differentiation [53,56,57]. HSCs are widely used for transplantation and gene therapy and there are 3 main sources of HSCs for HSC Transplantation (HSCT): UCB, BM, and PB [54,57]. Both HSCs and MSCs have been successfully used in the treatment of SSc [58].
MSCs in SSc
In recent years, the therapeutic efficacy of MSCs has been demonstrated in several preclinical animal models and is being evaluated in human clinical trials [59]. The roles of oxidative environment and crosstalk with other cells such as fibroblasts and endothelial cells on the functions of MSCs have been reported [59]. MSCs obtained from AT and BM have been used in the treatment of SSc using Intravenous (IV) and subcutaneous routes of administration [58-60].
Transplantation of a combination of autologous AT-MSCs and hyaluronic acid solution has been shown to cause significant improvement in the skin tightening without causing complications [60]. Autologous as well as allogeneic MSC transplantations have been used in the treatment of SSC [58,59]. MSCs could target the main features of the disease: vascular system (vasculopathy), tissue fibrosis, and immune system abnormalities [58]. The proangiogenic, immunomodulatory, anti-inflammatory, and immunosuppressive properties of MSCs make them ideal candidates for targeted cell-based therapies in SSc to restore the functions of the immune system [58,59]. The main outcomes of the clinical trials performed in patients with SSC include [59]:
- Improvement in perfusion, Raynaud’s phenomenon, and mouth opening
- Improvement in skin elasticity
- Reduction in pain
- Reduction in skin ulcerations
In patients with SSc, the combination of plasmapheresis and allogeneic MSC transplantation could produce sustained benefit in terms of: improvement in lung function, improvement in radiological features, and significant reduction in serological markers including: anti-Scl 70 antibody titers [61].
Digital ulcers represent a frequent manifestation of vasculopathy in patients with SSc [62]. However, currently there is no proven therapeutic strategy to promote healing of these ulcers. The MANUS (Mesenchymal Stromal Cells for Angiogenesis and Neovasularization in Digital Ulcers of Systemic Sclerosis) is the first double blind randomized controlled trial to assess safety and potential efficacy of MSCs in digital ulcers. In this on-going clinical trial, the study group arm will receive intramuscular MSCs while the control group will receive placebo [62].
In patients with SSc, MSCs obtained from BM and AT show upregulation of specific micro-RNAs (miRNAs) and downregulation of other miRNAs and the involved miRNAs exhibit profibrotic behavior. So, the miRNA profile or signature of MSCs may play a possible therapeutic role in treating patients with SSc [63]. In a murine model, repeated exposure of hypochloride resulted in a multistage process that leads to skin fibrosis. Infusion of MSCs during the process of fibrogenesis may halt the fibrotic process due to the great plasticity of MSCs [64].
Although some functional properties of MSCs could be affected upon culture with the serum of patients, MSCs can adapt to the oxidative environment and exert their therapeutic effects [65]. Recent evidences suggest that Extracellular Vesicles (ECVs) and exosomes play a role in the 3 main pathogenetic aspects of SSc: immunity, vascular damage, and fibrosis. Exosomes, which can be used as biological carriers, may have a potential role in the diagnostics, prognostics, and therapeutics in patients with SSc [66]. MSCs obtained from patients with SSc retain considerable immunosuppressive properties and a normal ability to generate functional T regulatory cells. However, evidence of their senescence does not represent a limitation for their potential use in cellular therapy and regenerative medicine to target scleroderma [67].
In 2 patients with refractory, progressive SSC, the combination of; plasmapheresis, rituximab, and IV MSCs derived from UCB resulted in clinical improvement lasting for more than 1 year [68]. In experimental models, it has been shown that the activity of inducible nitrous oxide synthase plays a crucial role in the antifibrotic activity of MSCs [64].
Transplantation of MSCs results in transferring miRNAs to the stem cells of the recipient in order to ameliorate osteopenia by recruiting non-coding RNA pathways [69]. MSCs obtained from patients with SSc may still harbor or retain some disease-specific abnormalities such as: intrinsic deregulation of vascular smooth muscle and myelofibroblastic transformation [70].
MSCs in Chronic sclerodermatous GVHD
Four patients with sclerodermatous GVHD received MSCs that were expanded ex-vivo from unrelated donors and administered by intra-BM injection [71]. Results of the study showed [71]:
- Dramatic reversal of helper T-lymphocyte 1 (Th1) cells to Th2 cells i.e. increase in Th1 and decrease in Th2 cells
- Gradual improvement in symptoms in all 4 patients. The authors conclude that MSC therapy in chronic GVHD was safe and beneficial
A prospective multicenter study that included 81 patients with BO following allogeneic HSCT randomized into [72]:
- Group A: 49 patients receiving azithromycin, prednisolone and MSCs
- Group B: 32 patients receiving azithromycin and prednisolone without MSCs
Reponses were encountered in 71% of group A and 44% of group B patients. Three years Overall Survival (OS) was 70.6% in group A and 58.2% in group B. There were no significant differences in the rates of infections or leukemia relapse between the 2 groups of patients. The authors concluded that MSC therapy was safe and effective in the treatment of BO encountered following allogeneic HSCT and that clinical improvement was accompanied by significant increase in IL-10 producing CD5+ B cells [72].
HSCT in SSc
Allogeneic HSCT has been performed in patients with SSc, but due to the high toxicity and Treatment-Related Mortality (TRM) most centers have focused on autologous HSCT because of its significantly lower organ toxicity and TRM [73]. Three randomized controlled clinical trials comprising 250 patients and comparing autologous HSCT to cyclophosphamide in the treatment of patients with SSc showed superior outcomes; in terms of longer OS and Event-Free Survival (EFS), improvement in skin condition and lung function, and decreased rates of disease progression; in recipients of autologous HSCT compared to patients receiving Cyclophosphamide (CPM) alone [74-76]. Details of these 3 randomized clinical trials are shown in Table 5 [74-76]. The positive outcomes of autologous HSCT in the 3 randomized controlled clinical trials were confirmed by the results of systematic reviews and meta-analyses that included other similar studies. Hence, two years ago, the American Society of Blood and Marrow Transplantation (ASBMT) issued a position statement that recommended autologous HSCT as the standard of care for patients with severe SSc [77-79].
The Randomized Clinical Trials and Details of the Clinical Trials | ASTIS (Autologous stem cell transplantation international scleroderma) |
(American scleroderma stem cell versus immune suppression trial) | SCOT (Scleroderma: cyclophosphamide or transplantation) |
Authors, Journal, and Year of Publication | van Laar JM, et al. JAMA 2014 | Burt RK, et al. Lancet 2011 | Sullivan KM, et al. NEJM 2018 |
Type of trial, Centers and Country of Origin of Trial | Phase III randomized multicenter trial 29 centers in 10 countries The Netherlands | Phase II randomized single center USA | Phase III randomized multicenter [26 sites] USA |
Number of Patients in the Study and in the Study Arms | 156 Autologous HSCT (79 patients) versus CPM (77 patients) | 19 Autologous HSCT (10 patients) versus CPM (9 patients) | 75 Autologous HSCT (36 patients) versus CPM (39 patients) |
Conditioning Therapy and Graft | Lymphoablative conditioning [CPM, rabbit ATG]CD34+ cell selection | Lymphoablative conditioning [CPM and rabbit ATG]No graft manipulation or CD34+ cell selection | Myeloablative conditioning [CPM, ATG, and TBI]CD34+ cell selection |
Basis of the Clinical Trial | Based on experience with similar regimens at: Saint Louis hospital in France and Leiden University Medical Center in the Netherlands No cross over to HSCT if failure on control arm | Based on phase I experience with 10 systemic sclerosis patients treated at North-Western University in USA | Based on phase I study of 33 patients mostly performed at the Fred Hutchinson Cancer Center in USA No cross over to HSCT if failure is encountered on control arm |
Results | (1) 1 year: – Events: 13 (16.5%) in HSCT arm versus 10.4% in CPM arm – TRM: 8 patients in HSCT arm versus 0 patients in CPM arm.
(2) 2 years: 14 events (17.7%) in HSCT arm versus 14 events (18.2%) in CPM arm.
(3) 4 years: 15 events (19%) in HSCT arm versus 20 events (26%) in CPM arm. | (1) at 12 months: all patients in HSCT arm improved and none experienced disease progression., while none of CPM arm patients improved and 8 patients had disease progression so 7 of them were switched to HSCT. (2) at 24 months: 11 HSCT patients had improvement in skin and lung condition | (1) EFS: – At 54 months: 79% in HSCT arm versus 50% in CPM arm with p value of 0.02 – At 72 months: 74% in HSCT arm versus 47% in CPM arm with p value of 0.03 (2) OS at 72 months: 86% in HSCT arm versus 51% in CPM arm with p value of 0.02 (3) TRM: – At 54 months: 3% in HSCT arm versus 0.00 % in CPM arm – At 72 months: 6% in HSCT arm versus 0.00% in CPM arm |
Conclusions |
HSCT arm had superior EFS and OS compared to CPM arm.
| Non-myeloablative autologous HSCT improved skin condition and pulmonary function for up to 2 years | Myeloablative autologous HSCT achieved long-term benefits including improved EFS and OS in patients with scleroderma but at the cost of increased expected toxicity |
HSCT: Hematopoietic Stem Cell Transplantation; ATG: Antithymocyte Globulin; CPM: Cyclophosphamide; TBI: Total Body Irradiation; TRM: Treatment Related Mortality; EFS: Event Free Survival; OS: Overall Survival |
Table 5: Details of the randomized controlled clinical trials on autologous HSCT in systemic sclerosis.
In a single center retrospective comparison-case control from historical cohort, Del papa N, et al., compared autologous HSCT in patients with rapidly progressive cutaneous SSc to conventional therapy with 18 patients in HSCT arm and 36 patients in the conventional therapy arm [80]. Patients in the autologous HSCT arm had mobilization with granulocyte-colony stimulating factor and CPM. Peripheral stem cell collection was performed using leukapheresis. The product was enriched with CD34+ cells. Conditioning therapy was composed of: CPM and ATG in addition to hydration, mesna, and methylprednisolone. Infection prophylaxis was composed of: ciprofloxacin, acyclovir and fluconazole. The conventional therapy was composed of CPM, prednisolone or methylprednisolone, azathioprine, and methotrexate. The selection criteria for autologous HSCT included: active disease, severe skin involvement, non-severe heart involvement, and non-severe lung involvement. Results of the clinical trial showed:
- TRM less than 5.6%
- Significantly better outcomes in the HSCT arm compared to the conventional therapy arm in terms of OS, stabilization of lung function, as well as reduction in skin thickening and disease activity.
The authors concluded that autologous HSCT in rapidly progressive SSc had superior outcomes compared to conventional therapies [80].
Several clinical studies on autologous HSCT in patients with SSc have shown improvement in: skin condition, organ function particularly lung and vascular system, and long-term survival despite the toxicity and relatively high TRM encountered in some of these trials [81-87]. Complications of autologous HSCT in patients with SSc include [81-89]:
- TRM and organ toxicity particularly cardiotoxicity
- Gonadal failure
- Various infections
- Autoimmune diseases such as myasthenia gravis
According to European Blood and Marrow Transplantation data, in patients with severe SSc, non-myeloablative conditioning therapy followed by autologous HSCT has been shown to be effective as durable responses with acceptable TRM have been encountered in two thirds of patients [90]. Additionally, the ASBMT has recommended that SSc should be considered a standard of care indication for autologous HSCT based on the evaluation of high-dose chemotherapy followed by autologous HSCT for the treatment of SSc in: several observational and retrospective studies, at least 4 randomized controlled clinical trials, as well as several systematic reviews and meta-analyses [74-86,91,92].
Baseline evaluation prior to HSCT should include comprehensive medical history and physical examination; laboratory tests including complete blood count and differential, renal and hepatic profiles, viral and hepatitis screens, chest X-ray, electrocardiogram, echocardiogram; and cardiac magnetic resonance imaging [77,93]. Ideal candidates for autologous HSCT in patients with SSc include [78,91,94]:
- Non-smokers
- Non-responders to standard immunosuppressive therapies
- Patients with cutaneous SSc within 4-5 years from the diagnosis with mild to moderate organ involvement particularly interstitial lung disease
- Patients having limited cutaneous SSc with progressive visceral involvement
- Patients with high-risk SSc experiencing severe lung and diffuse skin involvement at an early stage of the disease
Exclusion criteria for autologous HSCT in patients with SSc include [77,93]:
- Age > 65 years
- Pregnancy
- Drug or alcohol abuse
- Active psychiatric illness
- Inability to provide informed consent
- Active infection such as human immunodeficiency virus, human T-cell lymphotropic virus types 1 and 2, and hepatitis B and C virus infections
- Malignancy, myelodysplastic syndrome, or serious hematological disorder
- End-stage organ failure or severe organ dysfunction particularly liver, heart, lungs, and kidneys
Mechanisms of action of autologous HSCT in patients with SSc are unknown. However, proposed mechanisms include: ablation or reduction of the aberrant immune cells, particularly those in the T-cell compartment, followed by re-establishment of immunological tolerance or reconstitution of a new immune system that is self-tolerant [77,78,94,95]. In patients with SSc, the mechanism of action of autologous HSCT may not be solely immunological [62]. Generally, immunological changes encountered after autologous HSCT in SSc do not necessarily reflect the clinical responses [62]. Profibrotic cytokines and even autoantibodies hardly appear to be influenced by autologous HSCT in patients with SSc [96].
Autologous HSCT induces long-lasting alterations in B-cell homeostasis or in the B-cell compartment such as the increase production of IL-10 [97]. Clinical improvement or response following autologous HSCT in patients with SSc is associated with coordinated thymic and bone marrow rebounds manifested by increase counts of the newly generated T and B regulatory cells encountered after HSCT [98]. In patients with SSc, unlike conventional therapies, autologous HSCT leads to normalization or significant alterations of disease-related molecular signatures such as: decrease in interferon-α, reduction in neutrophil module, and increase in cytotoxic/natural killer cell module [99,100]. Compared to unmanipulated autologous grafts, CD34+ selected autologous HSCT may produce favorable effects on skin fibrosis and lung function. Therefore, use of CD34+ selected autologous HSCT with high-dose CPM conditioning therapy may offer excellent outcomes [101,102]. In patients with SSc receiving autologous HSCT with purified CD34+ cells, Th1/Th2 ratio has been reported to be significantly increased for at least 3 years following autologous HSCT [101].
Conclusions and Future Directions
Compared to standard therapy, autologous HSCT in patients with SSC has the following benefits [80,88,91,93,103]:
- Significant improvement in organ function
- Significant improvement in the QoL
- Remarkable improvement in long-term survival
Autologous HSCT is an effective therapeutic modality in selected patients with early diffuse cutaneous SSc, despite the expected toxicities and the relatively high TRM due to major organ involvement by the disease [104]. Better selection of candidates for HSCT and the use of safer transplant conditioning regimens may improve the outcome of autologous HSCT in patients with SSc [105]. Autologous HSCT in SSc should be performed by expert multidisciplinary teams in specialized transplant centers in order to optimize the safety and efficacy of the procedure [73]. More clinical studies, preferably multicenter randomized controlled trials, should be performed on MSC transplantation in specific forms and complications of SSc. In addition, not only the doses and routes of administration, but also the timing of MSC transplantation as well as the other required therapies should be determined.
References
- Sierra-Sepúlveda A, Esquinca-González A, Benavides-Suárez SA, Sordo-Lima DE, Caballero-Islas AE, Cabral-Castañeda AR, et al. Systemic sclerosis pathogenesis and emerging therapies, beyond the fibroblast. Biomed Res Int. 2019;2019:4569826.
- Sobolewski P, Maślińska M, Wieczorek M, Łagun Z, Malewska A, Roszkiewicz M, et al. Systemic sclerosis – multidisciplinary disease: clinical features and treatment. Reumatologia. 2019;57(4):221-33.
- Shah AA, Wigley FM. My approach to the treatment of scleroderma. Mayo Clin Proc. 2013;88(4):377-93.
- Katsiari CG, Simopoulou T, Alexiou I, Sakkas LI. Immunotherapy of systemic sclerosis. Hum Vaccin Immunother. 2018;14(11):2559-67.
- Gomer RH. Circulating progenitor cells and scleroderma. Curr Rheumatol Rep. 2008;10(3):183-8.
- Ho YY, Baron M, Recklies AD, Roughley PJ, Mort JS. Cells from the skin of patients with systemic sclerosis secrete chitinase 3-like protein 1. BBA Clin. 2014;1:2-11.
- Zimmermann AF, Pizzichini MM. Update on etiopathogenesis of systemic sclerosis. Rev Bras Reumatol. 2013;53(6):516-24.
- Abbot S, Bossingham D, Proudman S, de Costa C, Ho-Huynh A. Risk factors for the development of systemic sclerosis: a systematic review of the literature. Rheumatol Adv Pract. 2018;2(2):41.
- Fett N. Scleroderma: nomenclature, etiology, pathogenesis, prognosis, and treatments: facts and controversies. Clin Dermatol. 2013;31(4):432-7.
- Korman BD, Criswell LA. Recent advances in the genetics of systemic sclerosis: toward biological and clinical significance. Curr Rheumatol Rep. 2015;17(3):21.
- Scaletti C, Vultaggio A, Maggi E, Romagnani S, Piccinni MP. Microchimerism and systemic sclerosis. Int Arch Allergy Immunol. 2001;125(3):196-202.
- Arnett FC, Gourh P, Shete S, Ahn CW, Honey RE, Agarwal SK, et al. Major histocompatibility complex (MHC) class II alleles, haplotypes and epitopes which confer susceptibility or protection in systemic sclerosis: analyses in 1300 Caucasian, African-American and Hispanic cases and 1000 controls. Ann Rheum Dis. 2010;69(5):822-7.
- Tan FK, Arnett FC. Genetic factors in the etiology of systemic sclerosis and Raynaud phenomenon. Curr Opin Rheumatol. 2000;12(6):511-9.
- Kitko CL, White ES, Baird K. Fibrotic and sclerotic manifestations of chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2012;18(1):46-52.
- Raju G, Haghshenas A, Frieri M, Anand P. Sclerodermatous GVHD after allogeneic bone marrow transplant: a review. AIMS Cell Tissue Eng. 2017;1(1):3-11.
- Chosidow O, Bagot M, Vernant JP, Roujeau JC, Cordonnier C, Kuentz M, et al. Sclerodermatous chronic graft-versus-host disease: Analysis of seven cases. J Am Acad Dermatol. 1992;26(1):49-55.
- Ferri C, Bernini L, Cecchetti R, Latorraca A, Marotta G, Pasero G, et al. Cutaneous and serologic subsets of systemic sclerosis. J Rheumatol. 1991;18(12):1826-32.
- Hinchcliff M, Varga J. Systemic sclerosis/scleroderma: a treatable multisystem disease. Am Fam Physician. 2008;78(8):961-8.
- Hachulla E, Launay D. Diagnosis and classification of systemic sclerosis. Clin Rev Allergy Immunol. 2011;40(2):78-83.
- Wollheim FA. Classification of systemic sclerosis. Visions and reality. Rheumatol. 2005;44(10):1212-6.
- Lawley TJ, Peck GL, Moutsopoulos HM, Gratwohl AA, Deisseroth AB. Scleroderma, Sjögren-like syndrome, and chronic graft-versus-host disease. Ann Intern Med. 1977;87(6):707-9.
- Gratwohl AA, Moutsopoulos HM, Chused TM, Akizuki M, Wolf RO, Sweet JB, et al. Sjögren-type syndrome after allogeneic bone-marrow transplantation Ann Intern Med. 1977;87(6):703-6.
- Mekori YA, Claman HN. Is graft-versus-host disease a reliable model for scleroderma? Ric Clin Lab. 1986;16(4):509-13.
- Fleming JN, Shulman HM, Nash RA, Johnson PY, Wight TN, Gown A, et al. Cutaneous chronic graft-versus-host disease does not have the abnormal endothelial phenotype or vascular rarefaction characteristic of systemic sclerosis. PLoS One. 2009;4(7):e6203.
- Zhou L, Askew D, Wu C, Gilliam AC. Cutaneous gene expression by DNA microarray in murine sclerodermatous graft-versus-host disease, a model for human scleroderma. J Invest Dermatol. 2007;127(2):281-92.
- Molés-Poveda P, Montesinos P, Sanz-Caballer J, de Unamuno B, Piñana JL, Sahuquillo A, et al. Sclerodermatous chronic graft-versus-host disease treated with imatinib: a dermatological perspective. Actas Dermosifiliogr. 2018;109(3):241-7.
- Inamoto Y, Storer BE, Petersdorf EW, Nelson JL, Lee SJ, Carpenter PA, et al. Incidence, risk factors, and outcomes of sclerosis in patients with chronic graft-versus-host disease. Blood. 2013;121(25):5098-103.
- McCormick LL, Zhang Y, Tootell E, Gilliam AC. Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J Immunol. 1999;163(10):5693-9.
- Zhang Y, McCormick LL, Desai SR, Wu C, Gilliam AC. Murine sclerodermatous graft-versus-host disease, a model for human scleroderma: cutaneous cytokines, chemokines, and immune cell activation. J Immunol. 2002;168(6):3088-98.
- Pawlak-Buś K, Schmidt W, Olejarz M, Czyż A, Komarnicki M, Leszczyński P. Autologous hematopoietic stem cell transplant for progressive diffuse systemic sclerosis: procedural success and clinical outcome in 5-year follow-up. Reumatologia. 2019;57(1):50-4.
- Wells AU, Steen V, Valentini G. Pulmonary complications: one of the most challenging complications of systemic sclerosis. Rheumatol. 2009;48(3):40-44.
- Forbes A, Marie I. Gastrointestinal complications: the most frequent internal complications of systemic sclerosis. Rheumatol. 2009;48(3):36-9.
- Hasan O, Jessar M, Ashar M, Noordin S, Ahmad T. Systemic sclerosis: Clinical manifestations, anesthetic and orthopedic considerations in a patient. Int J Surg Case Rep. 2018;42:24-8.
- Fischer A, Patel NM, Volkmann ER. Interstitial lung disease in systemic sclerosis: focus on early detection and intervention. Open Access Rheumatol. 2019;11:283-307.
- Khanna D, Tashkin DP, Denton CP, Renzoni EA, Desai SR, Varga J. Etiology, risk factors, and biomarkers in systemic sclerosis with interstitial lung disease. Am J Respir Crit Care Med. 2020;201(6):650-60.
- Kahan A, Coghlan G, McLaughlin V. Cardiac complications of systemic sclerosis. Rheumatol. 2009;48(3):45-8.
- McFarlane IM, Bhamra MS, Kreps A, Iqbal S, Al-Ani F, Saladini-Aponte C, et al. Gastrointestinal manifestations of systemic sclerosis. Rheumatol. 2018;8(1):235.
- Kodet O, Oreská S. Systemic sclerosis mimics. In: New insights into systemic sclerosis. Edited by Tomcik M. Intech Open. 2019.
- Tyndall A, Fistarol S. The differential diagnosis of systemic sclerosis. Curr Opin Rheumatol. 2013;25(6):692-9.
- Jendrek ST, Kahle B, Riemekasten G. Mimetics of systemic sclerosis. Z Rheumatol. 2019;78(1):14-23.
- Nashel J, Steen V. Scleroderma mimics. Curr Rheumatol Rep. 2012;14(1):39-46.
- Chengappa KG, Negi VS. Hematopoietic stem cell transplantation in systemic sclerosis. Indian J Rheumatol. 2017;12(1):218-826.
- Mendoza FA, Mansoor M, Jimenez SA. Treatment of rapidly progressive systemic sclerosis: current and futures perspectives. Expert Opin Orphan Drugs. 2016;4(1):31-47.
- Matucci-Cerinic M, Steen VD, Furst DE, Seibold JR. Clinical trials in systemic sclerosis: lessons learned and outcomes. Arthritis Res Ther. 2007;9(2):1-9.
- Menon S, Shailendra S, Renda A, Longaker M, Quarto N. An overview of direct somatic reprogramming: the ins and outs of iPSCs. Int J Mol Sci. 2016;17(1):141.
- Al-Anazi KA. Stem cell treatments may reshape the future of medical therapeutics. J Stem Cell Biol Transplant. 2016;1(1:1).
- Diecke S, Jung SM, Lee J, Ju JH. Recent technological updates and clinical applications of induced pluripotent stem cells. Korean J Intern Med. 2014;29(5):547-57.
- Singh VK, Kalsan M, Kumar N, Saini A, Chandra R. Induced pluripotent stem cells: Applications in regenerative medicine, disease modeling, and drug discovery. Front Cell Dev Biol. 2015;3:2.
- Al-Anazi KA. Induced pluripotent stem cells and their future therapeutic applications in hematology. Stem Cell Res Ther. 2015;5(258):2.
- Al-Anazi KA, Al-Anazi WK, Al-Jasser AM. The Rising Role of Mesenchymal Stem Cells in the Treatment of Various Infectious Complications. Mesenchymal and Induced Pluripotent Stem Cells. 2020.
- Al-Anazi KA. Update on mesenchymal and induced pluripotent stem cells. In: Update on mesenchymal and induced pluripotent stem cells. Intech Open. 2019.
- Kumar A, D’Souza SS, Thakur AS. Understanding the journey of human hematopoietic stem cell development. Stem Cells Int. 2019;2019:2141475.
- Szade K, Gulati GS, Chan CKF, Kao KS, Miyanishi M, Marjon KD, et al. Where hematopoietic stem cells live: the bone marrow niche. Antioxid Redox Signal. 2018;29(2):191-204.
- Nakajima-Takagi Y, Osawa M, Iwama A. Manipulation of hematopoietic stem cells for regenerative medicine. Anat Rec. 2014;297(1):111-20.
- Seita J, Weissman IL. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip Rev Syst Biol Med. 2010;2(6):640-53.
- Nakajima H. Guest editorial: Hematopoietic stem cells. Int J Hematol. 2017;106(1):16-7.
- Zhang P, Zhang C, Li J, Han J, Liu X, Yang H. The physical microenvironment of hematopoietic stem cells and its emerging roles in engineering applications. Stem Cell Res Ther. 2019;10(1):1-13.
- Peltzer J, Aletti M, Frescaline N, Busson E, Lataillade JJ, Martinaud C. Mesenchymal stromal cells-based therapy in systemic sclerosis: rational and challenges. Front Immunol. 2018;9:2013.
- 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.
- Scuderi N, Ceccarelli S, Onesti MG, Fioramonti P, Guidi C, Romano F, et al. Human adipose-derived stromal cells for cell-based therapies in the treatment of systemic sclerosis. Cell Transplant. 2013;22(5):779-95.
- Zhang H, Liang J, Tang X, Wang D, Feng X, Wang F, et al. Sustained benefit from combined plasmapheresis and allogeneic mesenchymal stem cells transplantation therapy in systemic sclerosis. Arthritis Res Ther. 2017;19(1):165.
- Van Rhijn-Brouwer FCC, Gremmels H, Fledderus JO, Schuurman AH, Bonte-Mineur F, Vonk MC, 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(8):e020479.
- Di Benedetto P, Panzera N, Cipriani P, Mastroiaco V, Tessitore A, Liakouli V, et al. Mesenchymal stem cells of systemic sclerosis patients, derived from different sources, show a profibrotic microRNA profiling. Sci Rep. 2019;9(1):7144.
- Maria ATJ, Rozier P, Fonteneau G, Sutra T, Maumus M, Toupet K, et al. iNOS activity is required for the therapeutic effect of mesenchymal stem cells in experimental systemic sclerosis. Front Immunol. 2018;9:3056.
- Fonteneau G, Bony C, Goulabchand R, Maria ATJ, Le Quellec A, Rivière S, et al. Serum-mediated oxidative stress from systemic sclerosis patients affects mesenchymal stem cell function. Front Immunol. 2017;8:988.
- Colletti M, Galardi A, De Santis M, Guidelli GM, Di Giannatale A, Di Luigi L, et al. Exosomes in systemic sclerosis: messengers between immune, vascular and fibrotic components? Int J Mol Sci. 2019;20(18):4337.
- Cipriani P, Di Benedetto P, Liakouli V, Del Papa B, Di Padova M, Di Ianni M, et al. Mesenchymal stem cells (MSCs) from scleroderma patients (SSc) preserve their immunomodulatory properties although senescent and normally induce T regulatory cells (Tregs) with a functional phenotype: implications for cellular-based therapy. Clin Exp Immunol. 2013;173(2):195-206.
- Wehbe T, Abi Saab M, Abi Chahine N, Margossian T. Mesenchymal stem cell therapy for refractory scleroderma: a report of 2 cases. Stem Cell Investig. 2016;3:48.
- Chen C, Wang D, Moshaverinia A, Liu D, Kou X, Yu W, et al. Mesenchymal stem cell transplantation in tight-skin mice identifies miR-151-5p as a therapeutic target for systemic sclerosis. Cell Res. 2017;27(4):559-77.
- Hegner B, Schaub T, Catar R, Kusch A, Wagner P, Essin K, et al. Intrinsic deregulation of vascular smooth muscle and myofibroblast differentiation in mesenchymal stromal cells from patients with systemic sclerosis. PLoS One. 2016;11(4):e0153101.
- Zhou H, Guo M, Bian C, Sun Z, Yang Z, Zeng Y, et al. Efficacy of bone marrow-derived mesenchymal stem cells in the treatment of sclerodermatous chronic graft-versus-host disease: clinical report. Biol Blood Marrow Transplant. 2010;16(3):403-12.
- Chen S, Zhao K, Lin R, Wang S, Fan Z, Huang F, et al. The efficacy of mesenchymal stem cells in bronchiolitis obliterans syndrome after allogeneic HSCT: A multicenter prospective cohort study. EBioMedicine. 2019;49:213-22.
- Sullivan KM, Shah A, Sarantopoulos S, Furst DE. Hematopoietic stem cell transplantation for scleroderma: effective immunomodulatory therapy for patients with pulmonary involvement. Arthritis Rheumatol. 2016;68(10):2361-71.
- Burt RK, Shah SJ, Dill K, Gheorghiade M, Schroeder J, Craig R, 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(9790):498-506.
- Van Laar JM, Farge D, Sont JK, Naraghi K, Marjanovic Z, Larghero J, et al. EBMT/EULAR scleroderma study group. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. JAMA. 2014;311(24):2490-8.
- Sullivan KM, Goldmuntz EA, Keyes-Elstein L, McSweeney PA, Pinckney A, Welch B, et al. SCOT study investigators. Myeloablative autologous stem-cell transplantation for severe scleroderma. N Engl J Med. 2018;378(1):35-47.
- Host L, Nikpour M, Calderone A, Cannell P, Roddy J. Autologous stem cell transplantation in systemic sclerosis: a systematic review. Clin Exp Rheumatol. 2017;35(106):198-207.
- Shouval R, Furie N, Raanani P, Nagler A, Gafter-Gvili A. Autologous hematopoietic stem cell transplantation for systemic sclerosis: a systematic review and meta-analysis. Biol Blood Marrow Transplant. 2018;24(5):937-44.
- Sullivan KM, Majhail NS, Bredeson C, Carpenter PA, Chatterjee S, Crofford LJ, et al. Systemic sclerosis as an indication for autologous hematopoietic cell transplantation: Position statement from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2018;24(10):1961-4.
- Del Papa N, Onida F, Zaccara E, Saporiti G, Maglione W, Tagliaferri E, et al. Autologous hematopoietic stem cell transplantation has better outcomes than conventional therapies in patients with rapidly progressive systemic sclerosis. Bone Marrow Transplant. 2017;52(1):53-8.
- Binks M, Passweg JR, Furst D, McSweeney P, Sullivan K, Besenthal 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(6):577-84.
- Farge D, Marolleau JP, Zohar S, Marjanovic Z, Cabane J, Mounier N, 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(3):726-39.
- Vonk MC, Marjanovic Z, Van den Hoogen FH, Zohar S, Schattenberg AV, Fibbe WE, et al. Long-term follow-up results after autologous haematopoietic stem cell transplantation for severe systemic sclerosis. Ann Rheum Dis. 2008;67(1):98-104.
- McSweeney PA, Nash RA, Sullivan KM, Storek J, Crofford LJ, Dansey R, et al. High-dose immunosuppressive therapy for severe systemic sclerosis: initial outcomes. Blood. 2002;100(5):1602-10.
- Nash RA, McSweeney PA, Crofford LJ, Abidi M, Chen CS, Godwin JD, et al. High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for severe systemic sclerosis: long-term follow-up of the US multicenter pilot study. Blood. 2007;110(4):1388-96.
- Henes JC, Schmalzing M, Vogel W, Riemekasten G, Fend F, Kanz L, et al. Optimization of autologous stem cell transplantation for systemic sclerosis: a single-center long-term experience in 26 patients with severe organ manifestations. J Rheumatol. 2012;39(2):269-75.
- Mascarenhas S. Emerging use of stem cell transplants for systemic sclerosis. Med Res Arch. 2016;4(6).
- Walker UA, Saketkoo LA, Distler O. Haematopoietic stem cell transplantation in systemic sclerosis. RMD Open. 2018;4(1):e000533.
- Deligny C, Clave E, Sibon D, Daikeler T, Keshmandt H, Carmagnat M, et al. New onset of myasthenia gravis after treatment of systemic sclerosis by autologous hematopoietic stem cell transplantation: sustained autoimmunity or inadequate reset of tolerance? Hum Immunol. 2010;71(4):363-5.
- Farge D, Passweg J, van Laar JM, Marjanovic Z, Besenthal C, Finke J, et al. EBMT/EULAR Registry. Autologous stem cell transplantation in the treatment of systemic sclerosis: report from the EBMT/EULAR Registry. Ann Rheum Dis. 2004;63(8):974-81.
- Young A, Khanna D. Systemic sclerosis: a systematic review on therapeutic management from 2011 to 2014. Curr Opin Rheumatol. 2015;27(3):241-8.
- Eyraud A, Scouppe L, Barnetche T, Forcade E, Lazaro E, Duffau P, et al. Efficacy and safety of autologous haematopoietic stem cell transplantation in systemic sclerosis: a systematic review of the literature. Br J Dermatol. 2018;178(3):650-8.
- Farge D, Burt RK, Oliveira MC, Mousseaux E , Rovira M, Marjanovic Z, 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(11):1495-1503.
- Khanna D, Georges GE, Couriel DR. Autologous hematopoietic stem cell therapy in severe systemic sclerosis: ready for clinical practice? JAMA. 2014;311(24):2485-7.
- Burt RK, Patel D, Thomas J, Yeager A, Traynor A, Heipe F, et al. The rationale behind autologous autoimmune hematopoietic stem cell transplant conditioning regimens: concerns over the use of total-body irradiation in systemic sclerosis. Bone Marrow Transplant. 2004;34(9):745-51.
- Henes JC, Wirths S, Kötter I. Autologous stem cell transplantation in systemic sclerosis. Z Rheumatol. 2016;75(8):762-9.
- Gernert M, Tony HP, Schwaneck EC, Gadeholt O, Schmalzing M. Autologous hematopoietic stem cell transplantation in systemic sclerosis induces long-lasting changes in B-cell homeostasis toward an anti-inflammatory B-cell cytokine pattern. Arthritis Res Ther. 2019;21(1):106.
- Arruda LCM, Malmegrim KCR, Lima-Júnior JR, Clave E, Dias JBE, Moraes DA, et al. Immune rebound associates with a favorable clinical response to autologous HSCT in systemic sclerosis patients. Blood Adv. 2018;2(2):126-41.
- Assassi S, Wang X, Chen G, Goldmuntz E, Keyes-Elstein L, Ying J, et al. Myeloablation followed by autologous stem cell transplantation normalises systemic sclerosis molecular signatures. Ann Rheum Dis. 2019;78(10):1371-8.
- Fleming JN, Nash RA, McLeod DO, Fiorentino DF, Shulman HM, Connolly MK, et al. Capillary regeneration in scleroderma: stem cell therapy reverses phenotype? PLoS One. 2008;3(1):e1452.
- Tsukamoto H, Nagafuji K, Horiuchi T, Mitoma H, Niiro H, Arinobu Y, et al. Analysis of immune reconstitution after autologous CD34+ stem/progenitor cell transplantation for systemic sclerosis: predominant reconstitution of Th1 CD4+ T cells. Rheumatol. 2011;50(5):944-52.
- Ayano M, Tsukamoto H, Mitoma H, Kimoto Y, Akahoshi M, Arinobu Y, 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(1):30.
- Van Laar JM, Naraghi K, Tyndall A. Haematopoietic stem cell transplantation for poor-prognosis systemic sclerosis. Rheumatol. 2015;54(12):2126-33.
- Naraghi K, van Laar JM. Update on stem cell transplantation for systemic sclerosis: recent trial results. Curr Rheumatol Rep. 2013;15(5):326.
- Spierings J, van Rhijn-Brouwer FCC, van Laar JM. Hematopoietic stem-cell transplantation in systemic sclerosis: an update. Curr Opin Rheumatol. 2018;30(6):541-7.
Article Type
Review Article
Publication History
Received Date: 27-07-2020
Accepted Date: 08-09-2020
Published Date: 18-09-2020
Copyright© 2020 by Al-Anazi KA, et al. All rights reserved. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Citation: Al-Anazi KA, et al. Stem Cell Therapies in Systemic Sclerosis: An Updated Review. J Reg Med Biol Res. 2020;1(2):1-21.
1 | Unknown etiology |
2 | Autoimmune disorder associated with connective tissue diseases |
3 | Genetic susceptibility |
4 | Environmental exposure: vinylchloride, and organic solvents |
5 | Silicone breast implants |
6 | Infectious agents: |
a | Cytomegalovirus |
b | Epstein Barr virus |
c | Parvovirus B-19 |
d | Lyme disease |
7 | Medications: bleomycin, cocaine and pentazocine |
8 | Trauma |
9 | Radiation |
10 | Chronic graft versus host disease following allogeneic hematopoietic stem cell transplantation |
Table 1: Etiology and associations of systemic sclerosis.
1
| Cardiovascular
| · Heart block, arrhythmias, and other conduction disturbances |
· Diastolic dysfunction, left ventricular and congestive cardiac failure | ||
· Myocarditis, cardiac fibrosis and valvular abnormalities | ||
· Pericardial effusions, vasospasm and ischemic heart disease | ||
· Raynaud’s phenomenon, digital ischemia and digital ulcers | ||
2
| Gastrointestinal
| · Gastroesophageal reflux disease with dysphagia and regurgitation |
· Gastric antral vascular ectasia | ||
· Barrett esophagus, esophageal strictures and carcinoma of esophagus | ||
· Small intestinal bacterial overgrowth and intestinal malabsorption | ||
· Chronic large intestinal pseudo-obstruction | ||
· Xerostomia, gastroparesis and delayed gastric emptying | ||
· Fecal incontinence due to loss of internal anal sphincter function | ||
3 | Genito-urinary | · Sexual dysfunction |
4
| Musculoskeletal
| · Muscle weakness and atrophy, myopathy and myositis |
· Puffy hands and fingers and shortening of distal phalanges | ||
· Flexion contractures and restriction of joint movement | ||
· Tendon friction rubs and tenosynovitis | ||
· Arthritis and erosive arthropathy | ||
5
| Pulmonary
| · Pulmonary fibrosis, interstitial pneumonitis and interstitial lung disease |
· Pulmonary arterial hypertension and restrictive ventilator defects | ||
· Pulmonary infiltrates and ground-glass opacities on chest X-ray | ||
6 | Renal | · Renal crisis, proteinuria and azotemia |
7
| Skin
| · Calcinosis, shortening of distal phalanges. telangiectasia, pruritis and thick skin tags |
· Hypopigmentation and hyperpigmentation | ||
· Tight skin leading to: mask-line face and narrowing of mouth aperture | ||
8
| Central Nervous System | · Transient ischemic attack and stroke |
· Headache, dizziness, aphasia and visual disturbances | ||
· Anxiety, depression, convulsions and coma |
Table 2: Systemic complications of systemic sclerosis.
S. No. | Differential Diagnosis |
1 | Lichen sclerosis |
2 | Morphea form of basal cell carcinoma |
3 | Post-radiation morphea |
4 | Vaccination-associated morphea |
5 | Injection of silica, paraffin and vitamin-K |
6 | Esinophilic fascitis |
7 | Porphyria |
8 | Amyloidosis |
9 | Lipodermatosclerosis |
10 | Diabetic cheiroarthropathy/cheiroarthritis |
11 | Scleromyxedema |
12 | Nephrogenic systemic fibrosis |
13 | Mucinosis associated with thyroid disorders |
14 | Scleroderma adultorum Buschke |
Table 3: Differential diagnosis of systemic sclerosis.
1
| Treatment of Symptoms and Complications
| · Antimicrobials for infections |
· Proton pump inhibitors for gastrointestinal complications | ||
· Angiotensin-converting enzyme inhibitors and calcium channel blockers for hypertension | ||
2 | Immunosuppressive Therapies | · Cyclophosphamide, azathioprine, methotrexate and mycophenolate mofotil |
3
| Biological Therapies
| · Intravenous immunoglobulins and abatacept |
· Monoclonal antibodies such as: rituximab, tocilizumab, belimumab, inebilizumab and fresolimumab | ||
· Systemic targeted therapies including | ||
· Rapamycin | ||
· Tyrosine kinase inhibitors: imatinib, dasatinib, nilotinib | ||
4 | Stem Cell and Organ Transplantation | · Autologous hematopoietic stem cell transplantation for early diffuse systemic sclerosis |
Table 4: Available therapeutic modalities for systemic sclerosis.
The Randomized Clinical Trials and Details of the Clinical Trials | ASTIS (Autologous stem cell transplantation international scleroderma) |
(American scleroderma stem cell versus immune suppression trial) | SCOT (Scleroderma: cyclophosphamide or transplantation) |
Authors, Journal, and Year of Publication | van Laar JM, et al. JAMA 2014 | Burt RK, et al. Lancet 2011 | Sullivan KM, et al. NEJM 2018 |
Type of trial, Centers and Country of Origin of Trial | Phase III randomized multicenter trial 29 centers in 10 countries The Netherlands | Phase II randomized single center USA | Phase III randomized multicenter [26 sites] USA |
Number of Patients in the Study and in the Study Arms | 156 Autologous HSCT (79 patients) versus CPM (77 patients) | 19 Autologous HSCT (10 patients) versus CPM (9 patients) | 75 Autologous HSCT (36 patients) versus CPM (39 patients) |
Conditioning Therapy and Graft | Lymphoablative conditioning [CPM, rabbit ATG]CD34+ cell selection | Lymphoablative conditioning [CPM and rabbit ATG]No graft manipulation or CD34+ cell selection | Myeloablative conditioning [CPM, ATG, and TBI]CD34+ cell selection |
Basis of the Clinical Trial | Based on experience with similar regimens at: Saint Louis hospital in France and Leiden University Medical Center in the Netherlands No cross over to HSCT if failure on control arm | Based on phase I experience with 10 systemic sclerosis patients treated at North-Western University in USA | Based on phase I study of 33 patients mostly performed at the Fred Hutchinson Cancer Center in USA No cross over to HSCT if failure is encountered on control arm |
Results | (1) 1 year: – Events: 13 (16.5%) in HSCT arm versus 10.4% in CPM arm – TRM: 8 patients in HSCT arm versus 0 patients in CPM arm.
(2) 2 years: 14 events (17.7%) in HSCT arm versus 14 events (18.2%) in CPM arm.
(3) 4 years: 15 events (19%) in HSCT arm versus 20 events (26%) in CPM arm. | (1) at 12 months: all patients in HSCT arm improved and none experienced disease progression., while none of CPM arm patients improved and 8 patients had disease progression so 7 of them were switched to HSCT. (2) at 24 months: 11 HSCT patients had improvement in skin and lung condition | (1) EFS: – At 54 months: 79% in HSCT arm versus 50% in CPM arm with p value of 0.02 – At 72 months: 74% in HSCT arm versus 47% in CPM arm with p value of 0.03 (2) OS at 72 months: 86% in HSCT arm versus 51% in CPM arm with p value of 0.02 (3) TRM: – At 54 months: 3% in HSCT arm versus 0.00 % in CPM arm – At 72 months: 6% in HSCT arm versus 0.00% in CPM arm |
Conclusions |
HSCT arm had superior EFS and OS compared to CPM arm.
| Non-myeloablative autologous HSCT improved skin condition and pulmonary function for up to 2 years | Myeloablative autologous HSCT achieved long-term benefits including improved EFS and OS in patients with scleroderma but at the cost of increased expected toxicity |
HSCT: Hematopoietic Stem Cell Transplantation; ATG: Antithymocyte Globulin; CPM: Cyclophosphamide; TBI: Total Body Irradiation; TRM: Treatment Related Mortality; EFS: Event Free Survival; OS: Overall Survival |
Table 5: Details of the randomized controlled clinical trials on autologous HSCT in systemic sclerosis.