Next Article in Journal
Effect of Fenugreek on Hyperglycemia: A Systematic Review and Meta-Analysis
Next Article in Special Issue
Spontaneous Reversal of Vitiligo, a Rare Phenomenon Reported in a Case in Saudi Arabia with an Insight into Metabolic Biochemical Derangements
Previous Article in Journal
Management of Polydrug-Resistant Tuberculosis
Previous Article in Special Issue
Effect of Dupilumab on Sexual Desire in Adult Patients with Moderate to Severe Atopic Dermatitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Impact of Innovative Treatment Using Biological Drugs for the Modulation of Diffuse Cutaneous Systemic Sclerosis: A Systematic Review

by
Diego Fernández-Lázaro
1,2,*,
María Iglesias-Lázaro
1,
Evelina Garrosa
2,*,
Saray Rodríguez-García
3,4,
David Jerves Donoso
5,6,
Eduardo Gutiérrez-Abejón
7,8 and
Conrado Jorge-Finnigan
6,9
1
Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
2
Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
3
Department of Medicine, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42003 Soria, Spain
4
Internal Medicine Department of Soria University Assistance Complex (CAUSO), Santa Bárbara Hospital, Castile and Leon Health (SACyL), 42005 Soria, Spain
5
Pneumology Department of Soria University Assistance Complex (CAUSO), Santa Bárbara Hospital, Castile and Leon Health (SACyL), 42003 Soria, Spain
6
Department of Anatomy and Radiology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
7
Pharmacological Big Data Laboratory, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
8
Pharmacy Directorate, Castile and Leon Health Council, 47007 Valladolid, Spain
9
Dermatology Department of Soria University Assistance Complex (CAUSO), Santa Bárbara Hospital, Castile and Leon Health (SACyL), 42005 Soria, Spain
*
Authors to whom correspondence should be addressed.
Medicina 2023, 59(2), 247; https://doi.org/10.3390/medicina59020247
Submission received: 20 December 2022 / Revised: 19 January 2023 / Accepted: 26 January 2023 / Published: 27 January 2023
(This article belongs to the Special Issue Immune-Mediated Skin Diseases: Future Therapeutic Perspectives)

Abstract

:
Scleroderma or systemic sclerosis (SSc) is an autoimmune disease affecting the connective tissue, characterized by fibrosis of the skin and internal organs. There is currently no curative treatment available, so therapeutic action is aimed at a symptomatic treatment of the affected organs. The development of biotechnology has made it possible to implement certain biological drugs that could represent a window of opportunity to modulate the evolution and symptomatology of scleroderma with greater efficacy and less toxicity than conventional treatments. This study aimed to review the current evidence critically and systematically on the effects of biological drugs on the pulmonary function, skin disease, and health status of patients afflicted by diffuse cutaneous systemic sclerosis (dcSSc). Three electronic databases (Pubmed, Dialnet, and Cochrane Library Plus) were systematically searched until the cut-off date of October 2022. The review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and included original articles in English and Spanish with a controlled trial design, comparing biological drug treatments (tocilizumab, belimumab, riociguat, abatacept, and romilkimab) with a control group. The methodological quality of the studies was assessed using the McMaster quantitative form and the PEDro scale. A total of 383 studies were identified, 6 of them met the established criteria and were included in the present systematic review. A total of 426 patients treated with tocilizumab, belimumab, riociguat, abatacept, and romilkimab were included. The results showed substantial non-significant (p < 0.05) improvement trends after treatment with the biological drugs included in this review for the modified Rodnan Scale Value, Forced Vital Capacity, and Carbon Monoxide Diffusion Test; however, no benefits were shown on the Health Assessment Questionnaire–Disability Index when compared to the control group. Biological drugs, therefore, maybe a new therapeutic strategy for dcSSc and could be recommended as an additional and/or adjunctive treatment that promotes anti-fibrotic activity. This review could further define the clinical rationale for the use of biologics in the treatment of dcSSc and could provide key details on the study protocol, design, and outcome reporting.

1. Introduction

Scleroderma refers to a heterogeneous group of autoimmune fibrosing disorders. Etymologically, it is derived from Greek and means “hard skin” (skleros: hard; dermis: skin) [1]. Scleroderma was first described in 1752 by the Italian physician Carlo Curzio as a disease that “transformed the skin into wood” [2]. Generalized scleroderma or systemic sclerosis (SSc) is an autoimmune disease that affects the connective tissue, characterized by skin and organ fibrosis (heart, lungs, and kidneys), generalized microvasculopathy, and antibody responses against various cellular antigens and alterations in the immune system [3]. Clinically, two subtypes can be distinguished: (i) limited cutaneous systemic sclerosis (lcSSc), which progresses slowly by hardening skin in the acral areas, distal to elbows and/or knees, and on the face; (ii) diffuse cutaneous systemic sclerosis (dcSSc) which progresses rapidly by a thickening of the trunk and/or limb proximal regions (Table 1), [1]. Severe and serious affections occur in some organs (mainly in the lungs, heart, and kidneys), and has a poor prognosis [3]. In this way, Sulli et al. [4] demonstrated that blood perfusion is significantly lower in patients with SSc than in healthy subjects. The most affected parts were the fingertips, periungual, and palm areas, but not on the face or back of the hands, highlighting a selective affectation of the microcirculation due to the damage caused by SSc. These investigators used laser speckle contrast analysis (LASCA), which was an innovative safe technique to quantify blood perfusion (BP) in different areas of the body [4].
Currently, the annual incidence is 1 to 20 cases per million per year [10], with variations found depending on the geographical area, which may suggest that the rate of occurrence of new cases in the susceptible, at-risk population is conditioned by a genetic predisposition, individual hormonal behavioral determinants, and exposure to environmental factors [10,11,12]. The incidence is higher in the United States than in Europe, with a higher prevalence in African Americans than Caucasians [5]. SSc is more common in women than men, at a ratio of 3–5 women to every man, with the highest incidence rates between the ages of 30 and 50 years [5]. No gender differences were observed in vascular or gastrointestinal involvement; however, tendon rubbing or forced vital capacity (FVC) <70% occurs more commonly in men than in women. Regarding the age of mortality, it does not differ significantly between the sexes. Notwithstanding, for the male group, the development of symptoms and mortality is higher [12]. A positive correlation has been observed between different major histocompatibility complex (MHC) class II haplotypes and certain antibody subtypes (anti-centromere antibodies (ACA), HLA-DQB1*0501 and anti-topoisomerase) rarely found in healthy people or other connective tissue diseases [13,14]. Similarly, several alleles (HLA-DRB1*1501, DRB1*0701, DQA1*0102, DQB1*0602) have been described that could have a protective effect against SSc, as they have been found to be decreased in patients with the disease. The same polymorphism in the PTPN22 gene, associated with other autoimmune diseases, is also linked to SSc [15]. Exposure to silica, vinyl chloride, resins, and organic solvents and infections, cytomegalovirus, and parvovirus B19 are all known to trigger SSc in susceptible individuals [5].
The preliminary classificatory criteria for the classification of SSc were developed in 1980 by the American College of Rheumatology (ACR); advances in diagnostic tools and a joint effort by the ACR and the European League Against Rheumatism (EULAR) in 2013 allowed them to be expanded, giving them greater sensitivity and specificity, by including patients with early disease or very limited skin involvement (Table 2) [16]. In relation to laboratory findings, more than 95% of patients with SSc develop positive antinuclear antibodies (ANA), with ACA and anti-polymerase I being the most frequent, although up to seven specific ANA have been described; with anti-Scl70, anti-Th/To, and anti-U3RNP being those associated with a worse patient prognosis [17]. Other complementary tests include: radiodiagnostics for the detection of calcinosis; respiratory function tests assessing (FVC and carbon monoxide diffusion test (DLCO)); Doppler echocardiograms, electrocardiograms, and a Holter monitor for the study of cardiac involvement; barium esophageal study, esophageal manometries, breath tests and digestive endoscopies for digestive tract involvement; in addition, arterial monitoring and a Doppler ultrasound are used to assess renal involvement [18,19].
There is currently no curative or disease-modifying treatment available. Moreover, the loss of efficacy over time over long periods of treatment and with significant side effects has been demonstrated. Thus, therapeutic action will be aimed at symptomatic treatments of the affected organs according to their severity, depending on the evolution and duration of the disease [20]. Therefore, the need for an individualized precision pharmacological regimen seems obvious. To this end, therapeutic algorithms have been developed that provide for general measures such as first-, second-, and third-line drug regimens (Table 3) [20,21]. The Janus kinase (JAK) signaling pathway is an area of emerging interest in dermatology. In fact, recently, Moriana et al. [22] reported that JAK inhibitors could represent a safe and effective treatment option for cutaneous fibrosis and interstitial lung disease (ILD) in SSc. Additionally, the combination of two immunosuppressants, rituximab plus methotrexate, had potential efficacy for the skin and stabilization of internal organ involvement, and on some microangiopathies in early dcSSc [3]. In recent decades, the development of biotechnology has made it possible to implement hundreds of biological drugs, which are large protein molecules produced by living organisms that modulate the course of a disease by acting on a specific target. Biological drugs include hormones, monoclonal antibodies, blood products, immunomodulators, and vaccines [23]. According to the European Medicines Agency (EMA), a biosimilar is a biological drug that contains a version of the active substance of an original biological product or reference product whose patent has expired [24]. Biological drugs may represent a window of opportunity to modulate the progression and symptomatology of SSc with greater efficacy and lower toxicity than conventional treatments [20] (Table 4). Therefore, the purpose of this study was to review the current evidence critically and systematically on the effects of biological drugs on the health biomarkers of patients afflicted by dcSSc. The review protocol is published in the Prospective Registry of Systematic Reviews (PROSPERO); reference CRD42023387373.

2. Materials and Methods

2.1. Search Strategy

This systematic review was conducted following the specific methodological guidelines of the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) [26] and the PICO question model for the definition of the inclusion criteria: P (population): “dcSSc’s patients”; I (intervention): “treatment with biological drugs”; C (comparison): “same conditions with placebo, sham therapy or no intervention or pre/post comparison data group”; O (outcomes): “Skin disease (modified Rodnan scale value [mRSS]); pulmonary function test (forced vital capacity [FVC] and carbon monoxide diffusing capacity [DLCO]); and health status (Health Assessment Questionnaire [HAQ] Disability Index [DI] → HAQ-DI Scale). These parameters were included as outcomes as they are commonly investigated in health biomarker studies and in SSc research [27].
A structured search was carried out in the electronic databases: Medline (PubMed), Dialnet, and Cochrane Library Plus between September 2022 and December 2022. Publications from the last 5 years were included, given the evolution of the research in biological treatments for autoimmune diseases. Search terms included a mix of medical subject headings (MeSH) and free text words for key concepts related to biological drugs and SSc: scleroderma, systemic sclerosis, scleroderma diffuse, diffuse cutaneous systemic sclerosis, therapeutics, tocilizumab, biological therapy, antibodies, and monoclonal (monoclonal antibodies); all linked using the Boolean operators OR and AND. The complete search strategy is included in Appendix A. The review was carried out completely independently —titles, abstracts, and full texts—by two investigators (D.F.-L. and M.I.-L.). In addition, the inclusion criteria were independently evaluated and the disagreements generated were resolved by another reviewer (C.J-F.). There were no additional records of reference lists of the relevant articles or gray literature.

2.2. Inclusion and Exclusion Criteria

To select the studies, the following inclusion criteria were applied: (i) adults with the condition dcSSc; (ii) studies evaluating the effect of biological drugs accepted for the treatment of dcSSc in humans (excluding animal and/or in vitro studies); (iii) clinical trials, randomized and non-randomized trials, and pre-test/post-test designed studies (excluding reviews, notes, and other-than-original studies); (iv) studies evaluating outcomes (primary or secondary) of skin, respiratory, and functional capacity biomarkers; (v) studies that clearly report the dose, frequency, and route of drug administration; (vi) languages were restricted to English and Spanish; (vii) articles of methodological quality ≥ 11 points according to the McMaster University Occupational Therapy Evidence-Based Practice Research Group for quantitative studies [28] and ≥9 points according to the Physiotherapy Evidence Database (PEDro) scale [29]. Records that did not meet the criteria were excluded from this systematic review.

2.3. Methodological Quality Assessment

The methodological quality of the articles was assessed using the McMaster University Occupational Therapy Evidence-Based Practice Research Group [28] and the PEDro scale [29] as tools designed to assess the methodological quality of clinical designs.

2.4. Data Extraction

The data of the selected studies were summarized in Table 5. The following information was included: name of the first author, year of publication, country where the study was conducted, study design, sample size, gender and age of the participants, duration of the intervention, dose, and mode of administration of the treatment. This was performed by two study investigators (D.F.-L. and M.I.-L.) and disagreements were resolved by the intervention of another study investigator (C.J.-F.).

3. Results

3.1. Selection of Studies

We initially identified a total of 383 records. Among them, 20 duplicates were eliminated, 307 were not selected by study type, and 56 were not related to the objective of the study (systemic scleroderma). Two articles were also excluded after a full text review. The reasons for the exclusions after the full text review were inadequate results, because these studies were not related to the results evaluated in this study as skin disease, pulmonary function, and health status, and the remaining six studies [30,31,32,33,34,35] met our inclusion criteria and were included in the present systematic review (Figure 1).

3.2. Methodological Quality Assessment

One study was considered of “good quality” [31], one of “very good quality” [32], and four studies were considered of “excellent quality” [30,33,34,35], according to McMaster [28] (Table A1) For the PEDro scale [29], the score was 10 points for two studies [32,33] and 11 points for four studies [30,31,34,35], corresponding to “very good” and “excellent” quality, respectively (Table A2). No study was excluded for not reaching the minimum quality threshold (Table A1 and Table A2).

3.3. Characteristics of the Participants and Interventions

The total number of dcSSc patients included at the baseline in the studies was 426. All participants were ≥18 years old diagnosed with dcSSc according to the 1980 ACR or 2013 ACR/EULAR diagnostic criteria [16], with an active disease. In the review, we included six studies [30,31,32,33,34,35] that analyzed biological drugs: tocilizumab [32,35], with 2 different doses as 162 mg/week [32] and 8 mg/kg/month [35]; belimumab (10 mg/kg + 1000 mg twice/day mycophenolate mofetil [31]; riociguat (0.5–2.5 mg 3 times a day) [34]; abatacept (125 mg once a week) [33]; and romilkimab (200 mg/week) [30]. No study included patients of more than 5 years of evolution since the first symptom, excluding Raynaud’s phenomenon (RP) (Table 5).
One study excluded patients who had previously received mycophenolate mofetil > 3 months, rituximab or belimumab; or if they required >10 mg/day of prednisone [31]. Khanna et al. [34] excluded patients receiving nitrates, phosphodiesterase inhibitors, or SSc-specific treatments. Additionally, Shima et al. [35] excluded all patients who had used any biological drug in the previous 6 months. In the abatacepts study conducted by Khanna et al. [33], patients receiving immunomodulatory therapy were excluded. However, Allonote et al. [30] included patients on stable low-dose immunosuppressive therapy plus romilkimab.

3.4. Outcome Evaluation

A schematic summary was made of the different studies selected, organizing it into author(s) and year, study design, population, intervention, parameters analyzed, and main conclusions (Table 5).

3.5. Measure for Skin Disease

All studies (30–35) included in this systematic review evaluated the effectiveness of biological drugs on skin fibrosis using the mRSS scale. Only romilkimab [30] showed a significant decrease (p < 0.05) in induration compared to the control group. Tocilizumab [32,35], belimumab [31], riociguat [34], and abatacept [33] showed notable trends in the improvement on skin hardening.

3.6. Pulmonary Function Test

Lung functioning was assessed by FVC (30–34) and DLCO [30,31,32,34,35], with non-significant (p > 0.05) improvements in FVC being reported after the administration of belimumab [31], riociguat [34], romilkimab [30], and abatacept [33] when compared with the control group, and these increases in FVC were significant (p < 0.05) in belimumab-treated patients from the baseline to the end of the intervention [31]. DLCO showed a slight increase, yet non-significant (p > 0.05), in those patients treated with tocilizumab [32], belimumab [31], riociguat [34], and romilkimab [30] when compared to a control group. Belimumab treatment [31] showed significant (p < 0.05) increases in DLCO when compared with a baseline. On the contrary, Khanna et al. [32] reported non-significant (p > 0.05) decreases in the tocilizumab intervention group relative to the control and to the end of intervention after 48 weeks of treatment for both lung biomarkers FVC and DLCO.

3.7. Health Status

The alternative disability index assessed by the HAQ-DI scale was described in four studies included in this systematic review [30,32,33,34]. Treatments with riociguat [34], romilkimab [30], and abatacept [33] showed moderate non-significant decreases (p > 0.05) in the HAQ-DI scale with respect to the control group, while treatment with tocilizumab [32] did not induce any change when compared to the control group. Non-significant decreases (p > 0.05) also have been reported in the tocilizumab [32], romilkimab [30], and abatacept [33] intervention groups when comparing to the baseline. These results were contrary to those reported after 52 weeks of treatment with riociguat [34].

3.8. Safety

Adverse events were less reported in the intervention group compared to the control group in three studies (30,31,33), and more were reported in the tocilizumab and riociguat studies [32,34]. However, adverse events of severity were much lower in all five studies in the intervention group than in the control group. The most reported adverse effects in those patients were infections, followed by respiratory and gastrointestinal disorders (Table 6).

4. Discussion

The purpose of this systematic review was to critically study the impact of biological drug effects on the health biomarkers of patients afflicted by dcSSc. Six registers met the pre-specified inclusion/exclusion criteria. Overall, improvements have been reported regarding skin disease (mRSS) and pulmonary function parameters (FVC and DLCO), after treatment with the biological drugs included in this review. Yet, no benefits were shown on health status (HAQ-DI), and on the other hand, adverse effects associated with the use of biological drugs such as infections, respiratory, and gastrointestinal disorders may occur. Due to the differently measured results in the studies, the following results were divided into different sections to provide a clearer analysis.

4.1. Skin Disease

For a correct diagnosis, the thickening of the skin must be evaluated through the histological characteristics of the skin biopsy sample, adding subcutaneous tissue and muscle fascia. This type of skin biopsy allows analysis of the eosinophilic infiltrate, lymphocytes, and plasma cells [36]. However, the pathogenesis of dcSSc is complex and not completely understood, although there is evidence of the role of T and B lymphocytes in the production of profibrotic cytokines and in the activation of fibroblasts [37]. One of the cytokines implicated in the pathogenesis of dcSSc is interleukin 6 (IL-6), which is found in high concentrations in the skin and serum of patients with dcSSc [38], which in turn correlates with the severity of the disease due to the fact that IL-6 is related with high mRSS scores that represent the degree of skin involvement [38]. The results for treatment with tocilizumab [32,35] shows an improvement in dermal hardening. These results were consistent with those described by Denton et al. [39], which reported how protein production, migration, and contractility were reduced after 24 weeks of treatment with tocilizumab (162 mg) to an in vitro assay with dermal fibroblasts isolated from dcSSc patients, due to a blockade of interleukin-6 (IL-6) α-receptors.
The selective immunosuppressive action of Belimumab [31] on B lymphocytes significantly decreased skin hardening in dcSSc patients. This biological drug was also used to reduce skin hardening in patients with cutaneous lupus erythematosus [40]. The treatment with riociguat [34] and abatacept [33] also tended to decrease skin induration, while romilkimab [30] decreased dermal fibrosis significantly by binding and neutralizing IL-4 and IL-13, directly by fibroblast activation, and indirectly by stimulating transforming growth factor beta (TGF-β); adding on top of that, the direct effect of IL-4 on T-cell activation triggers fibroblast activation and drives collagen synthesis, as well as anti-topoisomerase-1 antibodies [41].
These results in skin alterations would encourage the use of biological drugs not only due to their capability of counteracting and balancing skin hardening, but stimulating non-fibrotic tissues in patients with dcSSc.

4.2. Pulmonary Involvement

Pulmonary disease is present in most patients with SSc and is currently the leading cause of mortality. Two types of damage predominate in these patients: interstitial lung disease and pulmonary arterial hypertension [42]. It is acknowledged that patients with SSc often demonstrate a restrictive pattern, with reduced forced FVC and DLCO. Therefore, the improvement of respiratory parameters is an indirect measure of the efficacy and of the pharmacological effect in SSc because it would reduce the thickening, rigidity, and scarring of the respiratory membrane, composed of the alveolar endothelium and the vascular endothelium, and the basement membrane that they both share [43]. In three studies [30,31,33] included in this systematic review with romilkimab [30], abatacept [33], and belimumab [31], there was a notable tendency to improve FVC and DLCO, while in one study of riociguat [34], the amelioration was only evidenced by FVC. The results of a network meta-analysis show that no treatment (cyclophosphamide, mycophenolate, cyclophosphamide plus high-dose prednisone, cyclophosphamide followed by azathioprine, rituximab, pirfenidone, nintedanib, and pomalidomide) influenced DLCO and only rituximab significantly decreased FVC compared to a placebo [44]. In addition, another study reported that cyclophosphamide plus azathioprine and mycophenolate did not significantly reduce the decline in FVC [45]. Improvements in DLCO would be especially important as it would indicate the restoration of the thickness of this lung membrane, which allows oxygen and carbon dioxide to pass through it, avoiding hypoxemia, which is one of the characteristic features of interstitial lung diseases, such as pulmonary fibrosis developed by dcSSc.
In addition, the benefits on DLCO could mean a restoration of blood flow problems, caused by an increase in the thickness of the respiratory membrane or a concomitant pulmonary vascular disease, which would improve the rate of gas transfer, by unblocking the circulation and increasing the opportunities for gas displacement. However, after the administration of tocilizumab [32,35], no benefits have been observed on pulmonary fusion monitoring parameters such as FVC or DLCO, results that are consistent with those obtained by Manfredi et al. [46] in rheumatoid arthritis with interstitial lung disease. Perhaps the decrease in IL-6 would be insufficient to stop fibrosis of the respiratory membrane. Therefore, romilkimab, abatacept, and belimumab would be the treatments with the potential to attenuate, modulate, and control respiratory disturbances in patients with dcSSc. This would position these drugs, due to the benefit they induce as biological therapies, as targeted anti-fibrotic treatments that would complete the traditional pharmacological management of combinations of immunosuppressants, particularly cyclophosphamide and mycophenolate mofetil.

4.3. Health Status

Potentially, the improvement in quality of life is based on the control of tissue fibrosis: (i) respiratory, because it would increase respiratory capacity; and (ii) dermal, which allows for a greater range of movement [47]. Symptomatic improvements would stimulate the performance of activities of daily living [48]. Although, in general, we have described in this review the mitigation of pulmonary and respiratory symptoms for patients treated with biological drugs, no HAQ-DI improvements have been obtained [30,31,32,33,34,35]. This could be because the improvements described are insufficient to improve the eight aspects of daily life during the last week (dressing and grooming, getting up, eating, walking, hygiene, reaching, grasping, and activities) assessed by the HAQ-DI [49]. Two important limitations must also be taken into account: the HAQ-DI is a self-perceptive health assessment questionnaire developed for patients with rheumatoid arthritis, whose adaptation for dcSSc has little correlation with the severity of the disease [46].

4.4. Safety

The use of biological therapies raises concerns due to the lack of knowledge about the long-term effects of blocking or the persistent inhibition of cytokines, or their receptors, or of immune cells that play a key role in their mechanisms [48]. Belimumab [31] was the biological treatment that induced the most adverse effects. These results are consistent with those reported after the administration of belimumab in patients with lupus erythematosus [50] with infections, especially of the upper respiratory tract, being the most common adverse effects. Comparatively, these side effects are less than the main adverse events of JAK inhibitors. Some of the side effects of JAK inhibitors were: infections, gastrointestinal disorders, elevated liver enzymes, and dyslipidemia, but they do not lead to the discontinuation of treatment [22].

4.5. Clinical Trials of Biological Drugs for the Modulation of Diffuse Cutaneous Systemic Sclerosis

Some clinical trials have been developed to show more evidence to incorporate biological drugs into a routine and/or care protocols. Roche Pharma (Basel, Switzerland) announced in 2021 that the US Food and Drug Administration (FDA) has approved Actemra/RoActemra® (tocilizumab) for subcutaneous administration to slow the rate of decline in lung function in adult patients with interstitial lung disease associated with SSc. Actemra/RoActemra was the first FDA-approved biologic therapy for the treatment of this disease. The FDA approval was based on data from the focusSced (ClinicalTrials.gov Identifier: NCT02453256.phase III) randomized, double-blind, placebo-controlled clinical trial conducted in 212 adults with SSc. Information from the fascinating (ClinicalTrials.gov Identifier: NCT01532869), phase II/III, randomized, double-blind, placebo-controlled clinical trial in patients with SSc was also used [44].
A recent clinical trial (ClinicalTrials.gov Identifier: NCT03844061), which will run until June 2024, at the Hospital for Special Surgery (New York, NY, USA) with the pharmaceutical GlaxoSmithKline (Bretford, UK) as a collaborator, is using a combination therapy of belimumab and rituximab (monoclonal antibody) for the treatment of SSc. It is hypothesized that a combination therapy of rituximab and belimumab with mycophenolate mofetil background therapy will improve SSc skin fibrosis compared to a placebo and mycophenolate mofetil treatment in a group of early SSc patients [51]. A randomized, double-blind, placebo-controlled phase II study to investigate the efficacy and safety of riociguat (Adempas, BAY63-2521) in patients with dcSSc (ClinicalTrials.gov Identifier: NCT02283762), sponsored by Bayer (Berlin, Germany) showed remarkable results in mRSS and the HAQ-DI [52].
Bristol-Myers Squibb (New York, NY, USA) launched a pilot study to evaluate the safety and efficacy of abatacept in patients with systemic sclerosis (ClinicalTrials.gov Identifier: NCT00442611) in collaboration with Stanford University (Stanford, CA, USA). The investigators have observed a marked clinical improvement after abatacept therapy in patients with SSc that was associated with the modulation of inflammatory pathways in the skin [53]. Despite the good preliminary results of romilkimab in relieving the symptoms of CSS and slowing its progression, on 9 January 2020, the European Commission granted orphan designation EU/3/19/2246 to Sanofi-Aventis Groupe, (Paris, France), for romilkimab for the treatment of SSc. Romilkimab was withdrawn from the Union Register for orphan drugs in March 2021 at the request of the sponsor [54].

4.6. Strengths and Limitations

The authors of this review acknowledge some limitations. First, a limited number of manuscripts met the inclusion criteria. Nevertheless, our systematic approach followed the PRISMA method [26] and the search was conducted using three electronic databases (Pubmed, Dialnet, and Cochrane Library Plus). The McMaster methodological quality assessment tool [28] and the PEDro scale [29] were used to ensure that all selected records met the minimum quality criteria, and a number of outcomes commonly used in chronic disease drug research were included. Further, our systematic review was registered in the PROSPERO (CRD42023387373) public database. Secondly, there is a large heterogeneity of studies in terms of outcomes, dosage of supplements, and duration of intervention that justifies caution in interpreting the results.

5. Conclusions

Biological drugs have the potential to provide a therapeutic alternative as an add-on and/or adjunctive treatment to biological drugs with anti-fibrotic activity. Considering the results included in this systematic review (MRSS, FVC, DLCO), biologics would potentially improve tissue function, highly correlated with survival, although no benefits over HAQ-DI have been reported, or severity of adverse effects. This review could further define the clinical rationale for the use of biologics in the treatment of dcSSc and could provide key details on the study protocol, design, and outcome reporting. However, further studies on the treatment of dcSSc with biologics are required.

Author Contributions

D.F.-L.: conceived and designed the research, analyzed, and interpreted the data, drafted the paper, and approved the final version submitted for publication; M.I.-L., E.G. and C.J.-F.: analyzed and interpreted the data and critically reviewed the paper; S.R.-G., D.J.D. and E.G.-A. prepared figures and/or tables and approved the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research has been funded by (i) Chair of Knowledge and Innovation “Caja Rural de Soria” (Spain) in the call for funding of research projects related to the completion of end-of-degree projects 2022, with project number SO-12-2022.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors want to thank: (i) Neurobiology Research Group, Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, for their collaboration on infrastructure computer support; and (ii) Blanca García Gómez, Director of the Chair of Knowledge and Innovation “Caja Rural de Soria” (Spain) for their support and involvement in this study.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A. Search Sequence Followed for Selection of Articles

#1 “Scleroderma, systemic sclerosis” [Mesh]
#2 “Scleroderma diffuse, diffuse cutaneous systemic sclerosis” [Mesh] #3 #1 OR #2
#4 “Therapeutics” [Mesh] OR “Tocilizumab” [Mesh] OR “Biological Therapy” [Mesh] OR “Antibodies, monoclonal” [Mesh] OR “Antibodies, monoclonal” [Mesh].
#5 #3 AND #4
The search was conducted in the following electronic databases: Medline (PubMed), Dialnet, and Cochrane Library Plus between September 2022 and December 2022. Languages were restricted to English and Spanish.
Table A1. Results of the methodological quality assessment of included studies—McMaster Critical Review Form for Quantitative Studies [28].
Table A1. Results of the methodological quality assessment of included studies—McMaster Critical Review Form for Quantitative Studies [28].
StudyItemTotal%Quality Score
12345678910111213141516
Allanore et al. (2020) [30]111111111111111116100E
Gordon et al. (2018) [31]11111111000010111168.7G
Khanna Dinesh et al.
(2018) [32]
11111111001111111487.5VG
Khanna et al. (2020) [33]11111111110111111593.7E
Khanna et al. (2020) [34]11111111110111111593.7E
Shima et al. (2019) [35]11111111111111011593.7E
Abbreviations: 0 = not fulfilled criterion; 1 = fulfilled criterion; E = excellent; VG = very good; G = good. Item 1: study purpose; item 2: literature review; item 3: study design; item 4: blinding; item 5: sample description; item 6: sample size; item 7: ethics and consent; item 8: validity of outcomes; item 9: reliability of outcomes; item 10: intervention description; item 11: statistical significance; item 12: statistical analysis; item 13: clinical importance; item 14: conclusions; item 15: clinical implications; and item 16: study limitations.
Table A2. Results of the methodological quality assessment of included studies—PEDro scale [29].
Table A2. Results of the methodological quality assessment of included studies—PEDro scale [29].
StudyItemTotal%Quality Score
1234567891011
Allanore et al. (2020) [30] 11100E
Gordon et al. (2018) [31] 11100E
Khanna Dinesh et al.
(2018) [32]
1090.9VG
Khanna et al. (2020) [33] 1090.9VG
Khanna et al. (2020) [34] 11100E
Shima et al. (2019) [35] 11100E
Abbreviations: Red = not fulfilled criterion; Green = fulfilled criterion; E = excellent; VG = very good; G = good; F = fair.

References

  1. Fett, N. Scleroderma: Nomenclature, etiology, pathogenesis, prognosis, and treatments: Facts and controversies. Clin. Dermatol. 2013, 31, 432–437. [Google Scholar] [CrossRef] [PubMed]
  2. Suliman, S.; Al Harash, A.; Roberts, W.N.; Perez, R.L.; Roman, J. Scleroderma-related interstitial lung disease. Respir. Med. Case Rep. 2017, 22, 109–112. [Google Scholar] [CrossRef] [PubMed]
  3. Smith, V.; Pizzorni, C.; Riccieri, V.; Decuman, S.; Brusselle, G.; de Pauw, M.; Deschepper, E.; Piette, Y.; Ruaro, B.; Sulli, A.; et al. Stabilization of Microcirculation in Patients with Early Systemic Sclerosis with Diffuse Skin Involvement following Rituximab Treatment: An Open-label Study. J. Rheumatol. 2016, 43, 995–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sulli, A.; Ruaro, B.; Cutolo, M. Evaluation of blood perfusion by laser speckle contrast analysis in different areas of hands and face in patients with systemic sclerosis. Ann. Rheum. Dis. 2014, 73, 2059–2061. [Google Scholar] [CrossRef] [PubMed]
  5. Carreira, P.E.; Martín-López, M.; Pablos Álvarez, J.L. Esclerodermia. Med.-Programa Form. 2017, 12, 1448–1457. [Google Scholar] [CrossRef]
  6. Khanna, D.; Furst, D.E.; Clements, P.J.; Allanore, Y.; Baron, M.; Czirjak, L.; Distler, O.; Foeldvari, I.; Kuwana, M.; Matucci-Cerinic, M.; et al. Standardization of the Modified Rodnan Skin Score for Use in Clinical Trials of Systemic Sclerosis. J. Scleroderma Relat. Disord. 2017, 2, 11–18. [Google Scholar] [CrossRef] [Green Version]
  7. Jaovisidha, K.; Csuka, M.; Almagro, U.A.; Soergel, K.H. Severe gastrointestinal involvement in systemic sclerosis: Report of five cases and review of the literature. Semin. Arthritis Rheum. 2005, 34, 689–702. [Google Scholar] [CrossRef]
  8. DeMarco, P.J.; Weisman, M.H.; Seibold, J.R.; Furst, D.E.; Wong, W.K.; Hurwitz, E.L.; Mayes, M.; White, B.; Wigley, F.; Barr, W.; et al. Predictors and outcomes of scleroderma renal crisis: The High-Dose Versus Low-Dose D-Penicillamine in early diffuse systemic sclerosis trial. Arthritis Rheum. 2002, 46, 2983–2989. [Google Scholar] [CrossRef]
  9. Steen, V.D.; Medsger, T.A., Jr. Severe organ involvement in systemic sclerosis with diffuse scleroderma. Arthritis Rheum. 2000, 43, 2437–2444. [Google Scholar] [CrossRef]
  10. Kuwana, M.; Saito, A.; Sakamoto, W.; Raabe, C.; Saito, K. Incidence Rate and Prevalence of Systemic Sclerosis and Systemic Scle-rosis-Associated Interstitial Lung Disease in Japan: Analysis Using Japanese Claims Databases. Adv. Ther. 2022, 39, 2222–2235. [Google Scholar] [CrossRef]
  11. De Almeida Chaves, S.; Porel, T.; Mounié, M.; Alric, L.; Astudillo, L.; Huart, A.; Lairez, O.; Michaud, M.; Prévot, G.; Ribes, D.; et al. Sine scleroderma, limited cutaneous, and diffused cutaneous systemic sclerosis survival and predictors of mortality. Arthritis Res. Ther. 2021, 23, 295. [Google Scholar] [CrossRef]
  12. Freire, M.; Rivera, A.; Sopeña, B.; Vilella, C.T.; Castillo, A.G.-D.; Argüelles, D.C.; Rubio, J.L.C.; Rivas, M.R.; Martínez, L.T.; Parra, J.A.T.; et al. Clinical and epidemiological differences between men and women with systemic sclerosis: A study in a Spanish systemic sclerosis cohort and literature review. Ann. Rheum. Dis. 2017, 35, 89–97. [Google Scholar]
  13. Stevens, A.M.; Kanaan, S.B.; Torok, K.S.; Medsger, T.A.; Mayes, M.D.; Reveille, J.D.; Klein-Gitelman, M.; Reed, A.M.; Lee, T.; Li, S.C.; et al. Brief Report: HLA-DRB1, DQA1, and DQB1 in Juvenile-Onset Systemic Sclerosis. Arthritis Rheumatol. 2016, 68, 2772–2777. [Google Scholar] [CrossRef]
  14. Yang, C.; Tang, S.; Zhu, D.; Ding, Y.; Qiao, J. Classical Disease-Specific Autoantibodies in Systemic Sclerosis: Clinical Features, Gene Susceptibility, and Disease Stratification. Front. Med. 2020, 7, 587773. [Google Scholar] [CrossRef]
  15. Diaz-Gallo, L.; Gourh, P.; Broen, J.; Simeon, C.; Fonollosa, V.; Ortego-Centeno, N.; Agarwal, S.; Vonk, M.; Coenen, M.; Riemekasten, G.; et al. Analysis of the influence of PTPN22 gene polymorphisms in systemic sclerosis. Ann. Rheum. Dis. 2010, 70, 454–462. [Google Scholar] [CrossRef] [Green Version]
  16. Van den Hoogen, F.; Khanna, D.; Fransen, J.; Johnson, S.R.; Baron, M.; Tyndall, A.; Matucci-Cerinic, M.; Naden, R.P.; Medsger, T.A., Jr.; Carreira, P.E.; et al. Classification Criteria for Systemic Sclerosis: An ACR-EULAR Collaborative Initiative. Arthritis Rheum. 2013, 65, 2737–2747. [Google Scholar] [CrossRef] [Green Version]
  17. Utsunomiya, A.; Oyama, N.; Hasegawa, M. Potential Biomarkers in Systemic Sclerosis: A Literature Review and Update. J. Clin. Med. 2020, 9, 3388. [Google Scholar] [CrossRef]
  18. Rutka, K.; Garkowski, A.; Karaszewska, K.; Łebkowska, U. Imaging in Diagnosis of Systemic Sclerosis. J. Clin. Med. 2021, 10, 248. [Google Scholar] [CrossRef]
  19. Gigante, A.; Leodori, G.; Pellicano, C.; Villa, A.; Rosato, E. Assessment of kidney involvement in systemic sclerosis: From scleroderma renal crisis to subclinical renal vasculopathy. Am. J. Med. Sci. 2022, 364, 529–537. [Google Scholar] [CrossRef]
  20. McMahan, Z.H.; Volkmann, E.R. An update on the pharmacotherapeutic options and treatment strategies for systemic sclerosis. Expert Opin. Pharmacother. 2020, 21, 2041–2056. [Google Scholar] [CrossRef]
  21. Codina, A.F.; Walker, K.M.; Pope, J.E. The Scleroderma Algorithm Group Treatment Algorithms for Systemic Sclerosis According to Experts. Arthritis Rheumatol. 2018, 70, 1820–1828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Moriana, C.; Moulinet, T.; Jaussaud, R.; Decker, P. JAK inhibitors and systemic sclerosis: A systematic review of the literature. Autoimmun. Rev. 2022, 21, 103168. [Google Scholar] [CrossRef] [PubMed]
  23. AseBio. Innovative Biological Medicines and Biosimilars. Available online: https://www.asebio.com/areas-de-trabajo/salud/medicamentos-biologicos-biosimilares (accessed on 13 December 2022).
  24. European Medicines Agency. Information Guide for Healthcare Professionals Produced Jointly by the European Medicines Agency and the European Commission. Available online: https://www.ema.europa.eu/en/documents/leaflet/biosimilars-eu-information-guide-healthcare-professionals_es.pdf (accessed on 15 December 2022).
  25. Bruni, C.; Cuomo, G.; Rossi, F.W.; Praino, E.; Bellando-Randone, S. Kidney involvement in systemic sclerosis: From pathogenesis to treatment. J. Scleroderma Relat. Disord. 2018, 3, 43–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Page, M.J.; Moher, D.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. PRISMA 2020 explanation and elaboration: Updated guidance and exemplars for reporting systematic reviews. BMJ 2021, 372, n160. [Google Scholar] [CrossRef] [PubMed]
  27. Kowal-Bielecka, O.; Landewé, R.; Avouac, J.; Chwiesko, S.; Miniati, I.; Czirják, L.; Clements, P.; Denton, C.; Farge, D.; Fligelstone, K.; et al. EULAR recommendations for the treatment of systemic sclerosis: A report from the EULAR Scleroderma Trials and Research group (EUSTAR). Ann. Rheum. Dis. 2009, 68, 620–628. [Google Scholar] [CrossRef]
  28. Law, M.; Stewart, C.; Pollock, N.; Letts, L.; Bosch, J.; Westmorland, M. Guidelines for Critical Review of Qualitative Studies; McMaster University Occupational Therapy Evidence-Based Practice Research Group: Hamilton, ON, Canada, 1998; pp. 1–9. [Google Scholar]
  29. Maher, C.G.; Sherrington, C.; Herbert, R.D.; Moseley, A.M.; Elkins, M. Reliability of the PEDro Scale for Rating Quality of Randomized Controlled Trials. Phys. Ther. 2003, 83, 713–721. [Google Scholar] [CrossRef] [Green Version]
  30. Allanore, Y.; Wung, P.; Soubrane, C.; Esperet, C.; Marrache, F.; Bejuit, R.; Lahmar, A.; Khanna, D.; Denton, C.P. A randomised, double-blind, placebo-controlled, 24-week, phase II, proof-of-concept study of romilkimab (SAR156597) in early diffuse cutaneous systemic sclerosis. Ann. Rheum. Dis. 2020, 79, 1600–1607. [Google Scholar] [CrossRef]
  31. Gordon, J.K.; Martyanov, V.; Franks, J.M.; Bernstein, E.J.; Szymonifka, J.; Magro, C.; Wildman, H.F.; Wood, T.A.; Whitfield, M.L.; Spiera, R.F. Belimumab for the Treatment of Early Diffuse Systemic Sclerosis: Results of a Randomized, Double-Blind, Placebo-Controlled, Pilot Trial. Arthritis Rheumatol. 2018, 70, 308–316. [Google Scholar] [CrossRef] [Green Version]
  32. Khanna, D.; Denton, C.P.; Lin, C.J.F.; Van Laar, J.M.; Frech, T.M.; Anderson, M.E.; Baron, M.; Chung, L.; Fierlbeck, G.; Lakshminarayanan, S.; et al. Safety and efficacy of subcutaneous tocilizumab in systemic sclerosis: Results from the open-label period of a phase II randomised controlled trial (faSScinate). Ann. Rheum. Dis. 2017, 77, 212–220. [Google Scholar] [CrossRef]
  33. Khanna, D.; Spino, C.; Johnson, S.; Chung, L.; Whitfield, M.L.; Denton, C.P.; Berrocal, V.; Franks, J.; Mehta, B.; Molitor, J.; et al. Abatacept in Early Diffuse Cutaneous Systemic Sclerosis: Results of a Phase II Investigator-Initiated, Multicenter, Double-Blind, Randomized, Placebo-Controlled Trial. Arthritis Rheumatol. 2020, 72, 125–136. [Google Scholar] [CrossRef]
  34. Khanna, D.; Allanore, Y.; Denton, C.P.; Kuwana, M.; Matucci-Cerinic, M.; Pope, J.E.; Atsumi, T.; Bečvář, R.; Czirják, L.; Hachulla, E.; et al. Riociguat in patients with early diffuse cutaneous systemic sclerosis (RISE-SSc): Randomised, double-blind, placebo-controlled multicentre trial. Ann. Rheum. Dis. 2020, 79, 618–625. [Google Scholar] [CrossRef] [Green Version]
  35. Shima, Y.; Kawaguchi, Y.; Kuwana, M. Add-on tocilizumab versus conventional treatment for systemic sclerosis, and cytokine analysis to identify an endotype to tocilizumab therapy. Mod. Rheumatol. 2018, 29, 134–139. [Google Scholar] [CrossRef] [Green Version]
  36. Foti, R.; De Pasquale, R.; Bosco, Y.D.; Visalli, E.; Amato, G.; Gangemi, P.; Foti, R.; Ramondetta, A. Clinical and Histopathological Features of Scleroderma-like Disorders: An Update. Medicina 2021, 57, 1275. [Google Scholar] [CrossRef]
  37. Fang, D.; Chen, B.; Lescoat, A.; Khanna, D.; Mu, R. Immune cell dysregulation as a mediator of fibrosis in systemic sclerosis. Nat. Rev. Rheumatol. 2022, 18, 683–693. [Google Scholar] [CrossRef]
  38. Shima, Y. Cytokines Involved in the Pathogenesis of SSc and Problems in the Development of Anti-Cytokine Therapy. Cells 2021, 10, 1104. [Google Scholar] [CrossRef]
  39. Denton, C.P.; Ong, V.H.; Xu, S.; Chen-Harris, H.; Modrusan, Z.; Lafyatis, R.; Khanna, D.; Jahreis, A.; Siegel, J.; Sornasse, T. Therapeutic interleukin-6 blockade reverses transforming growth factor-beta pathway activation in dermal fibroblasts: Insights from the faSScinate clinical trial in systemic sclerosis. Ann. Rheum. Dis. 2018, 77, 1362–1371. [Google Scholar] [CrossRef]
  40. Fairley, J.; Oon, S.; Saracino, A.; Nikpour, M. Management of cutaneous manifestations of lupus erythematosus: A systematic review. Semin. Arthritis Rheum. 2020, 50, 95–127. [Google Scholar] [CrossRef]
  41. Fuschiotti, P. Role of IL-13 in systemic sclerosis. Cytokine 2011, 56, 544–549. [Google Scholar] [CrossRef]
  42. Navarro, C. Pulmonary involvement in systemic sclerosis. Alveolitis, fibrosis and pulmonary arterial hypertension. Reumatol. Clínica 2006, 2, S16–S19. [Google Scholar] [CrossRef] [PubMed]
  43. Distler, O.; Volkmann, E.R.; Hoffmann-Vold, A.M.; Maher, T. Current and future perspectives on management of systemic sclerosis-associated interstitial lung disease. Expert Rev. Clin. Immunol. 2019, 15, 1009–1017. [Google Scholar] [CrossRef] [Green Version]
  44. Erre, G.L.; Sebastiani, M.; Fenu, M.A.; Zinellu, A.; Floris, A.; Cavagna, L.; Renzoni, E.; Manfredi, A.; Passiu, G.; Woodman, R.J.; et al. Efficacy, Safety, and Tolerability of Treatments for Systemic Sclerosis-Related Interstitial Lung Disease: A Systematic Review and Network Meta-Analysis. J. Clin. Med. 2020, 9, 2560. [Google Scholar] [CrossRef] [PubMed]
  45. Zheng, J.-N.; Yang, Q.-R.; Zhu, G.-Q.; Pan, L.; Xia, J.-X.; Wang, Q. Comparative efficacy and safety of immunosuppressive therapies for systemic sclerosis related interstitial lung disease: A Bayesian network analysis. Mod. Rheumatol. 2019, 30, 687–695. [Google Scholar] [CrossRef] [PubMed]
  46. Manfredi, A.; Cassone, G.; Furini, F.; Gremese, E.; Venerito, V.; Atzeni, F.; Arrigoni, E.; Della Casa, G.; Cerri, S.; Govoni, M.; et al. Tocilizumab therapy in rheumatoid arthritis with interstitial lung disease: A multicentre retrospective study. Intern. Med. J. 2019, 50, 1085–1090. [Google Scholar] [CrossRef]
  47. Allanore, Y.; Collaborators, O.B.O.T.E.; Bozzi, S.; Terlinden, A.; Huscher, D.; Amand, C.; Soubrane, C.; Siegert, E.; Czirják, L.; Carreira, P.E.; et al. Health Assessment Questionnaire-Disability Index (HAQ-DI) use in modelling disease progression in diffuse cutaneous systemic sclerosis: An analysis from the EUSTAR database. Thromb. Haemost. 2020, 22, 1–11. [Google Scholar] [CrossRef]
  48. Pope, J. Measures of systemic sclerosis (scleroderma): Health Assessment Questionnaire (HAQ) and Scleroderma HAQ (SHAQ), Physician- and Patient-Rated Global Assessments, Symptom Burden Index (SBI), University of California, Los Angeles, Scleroderma Clinical Trials. Arthritis Care Res. 2011, 63, S98–S111. [Google Scholar] [CrossRef]
  49. Johnson, S.R.; Hawker, G.A.; Davis, A.M. The health assessment questionnaire disability index and scleroderma health assessment questionnaire in scleroderma trials: An evaluation of their measurement properties. Arthritis Rheum. 2005, 53, 256–262. [Google Scholar] [CrossRef]
  50. Guerreiro Castro, S.; Isenberg, D.A. Belimumab in systemic lupus erythematosus (SLE): Evidence-to-date and clinical usefulness. Ther. Adv. Musculoskelet. Dis. 2017, 9, 75–85. [Google Scholar] [CrossRef] [Green Version]
  51. ClinicalTrialsgov. Belimumab and Rituximab Combination Therapy for the Treatment of Diffuse Cutaneous Systemic Sclerosis. Available online: https://clinicaltrials.gov/ct2/show/NCT03844061 (accessed on 14 January 2023).
  52. Good Clinical Practice. Clinical trials on Systemic Scleroderma: Diffuse Scleroderma: Systemic Scleroderma: Riociguat: Riociguat (Adempas, BAY63-2521). Clinical Trials Registry. Available online: https://ichgcp.net/es/clinical-trials-registry/NCT02283762 (accessed on 15 January 2023).
  53. Chakravarty, E.F.; Martyanov, V.; Fiorentino, D.; Wood, T.A.; Haddon, D.J.; Jarrell, J.A.; Utz, P.J.; Genovese, M.C.; Whitfield, M.L.; Chung, L. Gene expression changes reflect clinical response in a placebo-controlled randomized trial of abatacept in patients with diffuse cutaneous systemic sclerosis. Thromb. Haemost. 2015, 17, 1–14. [Google Scholar] [CrossRef] [Green Version]
  54. European Medicines Agency. EU/3/19/2246. Available online: https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu3192246 (accessed on 15 January 2023).
Figure 1. Flow chart depicting the processes of identifying and selecting relevant studies according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.
Figure 1. Flow chart depicting the processes of identifying and selecting relevant studies according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.
Medicina 59 00247 g001
Table 1. Clinical signs, prevalence, and symptomatology in diffuse cutaneous systemic sclerosis (dcSSc).
Table 1. Clinical signs, prevalence, and symptomatology in diffuse cutaneous systemic sclerosis (dcSSc).
Involved Organ(s)Clinical SignsPrevalenceSymptomatology
Vascular [4,5]Raynaud’s phenomenon100%Changes in skin coloration
Digital ulcers41.6%Hard-to-heal lesions → infection,
osteomyelitis, gangrene
Cutaneous [6]Dermal hardening100%
Pigmentation alterationsPatchy areas of depigmentation.
Calcinosis cutisSubcutaneous nodules
Pain
Musculoskeletal [5]Fibrosis46–97%Contractures
Morning stiffness
ArthritisArthralgias
Gastrointestinal [7] Esophageal hypomotility90%Dysphagia
Gastroesophageal refluxBacterial overgrowth
Stomach hypomotilityDelayed emptying → Premature satiety, fullness, bloating, nausea
Intestinal hypomotilityChronic intestinal pseudo-obstruction
Malabsorption
Anorectal dysfunctionIncontinence
Renal [8]Renal scleroderma crisis10%Malignant high blood pressure
Angiopathic hemolytic anemia
Thrombocytopenia
Proteinuria
Macrohematuria
Acute renal failure
Cardiac [9]ArrhythmiasVentricular90%Fatigue
Palpitations
Syncope
Dizziness
Supraventricular66%
Ventricular multiform premature beats40%
Left bundle branch block16%
First-degree
atrioventricular blocks
8%
Pericardial
conditions
Pericardial effusion78%Chest pain
Dyspnea
Fever
Pericarditis77.5%
Myocardial
dysfunction
RV dysfunction69%Fatigue
Calf edema
Dyspnea
Venous congestion
LV dysfunction46%
Cardiac failure20–25%
Valvular dysfunctionMitral valves
prolapse
20%Dyspnea
Pain
Edema
Palpitations
Fatigue
Pulmonary [2]Pulmonary interstitial disease40%Dyspnea
Dry cough
Crackles
Pulmonary arterial hypertension
Cor pulmonale
Pulmonary arterial hypertension15–20%RV hypertrophy
↓ Cardiac output
Heart failure
Abbreviations: RV: right ventricle; LV: left ventricle; ↓: decrease; →: produces.
Table 2. ACR/EULAR 2013 Systemic Scleroderma classification criteria. Adapted from Van Den Hoogen et al. [16].
Table 2. ACR/EULAR 2013 Systemic Scleroderma classification criteria. Adapted from Van Den Hoogen et al. [16].
ItemsSub-Item(s)Weight/Score
Skin thickening of the fingers of both hands extending proximal to the metacarpophalangeal joints (sufficient criterion) 9
Skin thickening of the fingers (only count the higher score)Puffy fingers2
Sclerodactyly of the fingers (distal to the metacarpophalangeal joints but proximal to the interphalangeal joints)4
Fingertip lesions (only count the higher score)Digital tip ulcers2
Fingertip pitting scars3
Telangiectasia 2
Abnormal nailfold capillaries 2
Pulmonary arterial hypertension and/or interstitial lung disease (maximum score is 2)Pulmonary arterial
hypertension
2
Interstitial lung disease2
Raynaud’s phenomenon 2
SSc-related autoantibodies (anticentromere, anti-topoisomerase I [anti–Scl-70], anti-RNA polymerase III) (maximum score is 3)Anticentromere3
Anti–topoisomerase I3
Anti–RNA polymerase III3
Abbreviations: SSc: systemic sclerosis. These criteria are applicable to any patient considered for inclusion in an SSc study. The criteria are not applicable to patients with skin thickenings paring the fingers or to patients who have a scleroderma-like disorder that better explains their manifestations (e.g., nephrogenic sclerosing fibrosis, generalized morphea, eosinophilic fasciitis, scleredema diabeticorum, scleromyxedema, erythromyalgia, porphyria, lichen sclerosis, graft-versus-host disease, diabetic cheerio arthropathy). The total score is determined by adding the maximum weight (score) in each category. Patients with a total score of 9 are classified as having definite SSc.
Table 3. Therapeutic strategies used in the treatment of systemic sclerosis (SSc).
Table 3. Therapeutic strategies used in the treatment of systemic sclerosis (SSc).
AffectationGeneral Measures1st Line Treatment2nd Line Treatment3rd Line TreatmentNew Therapeutic Tools
Peripheral vascular [4,5]Avoid exposure to cold, sudden changes in
temperature, stress, smoking,
vasospastic
substances.
Calcium channel
blockers
Phosphodiesterase-5
inhibitors
If severe RP:
Prostanoids
Prostacyclins and prostaglandins (Iloprost, Treprostanil)
Statins
Topical agents
Biologic drugs (Riociguat, Bosentan)
Botulinum toxin
If mild RP: Calcium channel blockers or
angiotensin receptor blockers.
Skin involvement
Skin [5]mRSS ≤ 32Avoid friction and traumaMethotrexateMycophenolate mofetil Pirfenidone
Nintendanib
Rituximab
mRSS > 32Mycophenolate mofetilMethotrexateCyclophosphamidesHematopoietic cell transplantation
Pirfenidone
Nintendanib
Rituximab
DU preventionCalcium channel
blockers
Phosphodiesterase-5
inhibitors
Bosentan
Prostaglandins
DU treatmentCalcium channel
blockers
Phosphodiesterase-5
inhibitors
Prostanoids
Musculoskeletal [5]Moderate exercise and physiotherapyMethotrexateLow dose Corticosteroid HydroxychloroquinesRituximab
Tocilizumab
Abatacept
Gastrointestinal [7]↓ MotilityPrevent
malnutrition
Proton pump
inhibitors
Agents that high motilitySurgical treatment
Bacterial
overgrowth
Antibiotherapy
Renal [25] Angiotensin-converting enzyme inhibitorsCalcium channel blockers
Aldosterone II receptor antagonists
Renal replacement
therapy
Dialysis
Renal Crisis [25] Renal replacement
therapy
Dialysis
Cardiac [9]Arrhythmias Verapamil
Amiodarone
Ablation
Pacemaker
Pericardial
involvement
Anti-inflammatory drugs
Corticosteroids
Pericardial drainage
Pericardiocentesis
Myocardial
dysfunction
Angiotensin-converting enzyme inhibitors
Beta-blockers
Diuretics
Cardiac resynchronization
Valvular
dysfunction
Cardiac transplantation
Transplantation of affected valves
Pulmonary [2] Mycophenolate
mofetil
Lung transplantationRituximab
Tocilizumab
Abbreviations: RP: Raynaud’s phenomenon; MmRSS: modified Rodnan scale; DU: digital ulcers.
Table 4. Function and mechanism of the biological drugs used for the treatment of systemic sclerosis (SSc) included in this systematic review.
Table 4. Function and mechanism of the biological drugs used for the treatment of systemic sclerosis (SSc) included in this systematic review.
DrugFunctionMechanism
TocilizumabImmunosuppressantIt binds to soluble and membrane-bound IL-6 receptors and inhibits IL-6-mediated signaling
BelimumabSelective immunosuppressantIgG1 monoclonal antibody that specifically binds to the soluble form of human B-cell-stimulating protein
RiociguatAnti-hypertension pulmonaryStimulator of the soluble guanylyl cyclase
AbataceptSelective immunosuppressantSelectively modulates a key co-stimulatory signal for the full activation of T lymphocytes expressing CD28
RomilkimabSelective immunosuppressantBiospecific antibody to IgG4 which neutralizes IL-4 and IL-13
Abbreviations: IgG: immunoglobulin G; IL: interleukin.
Table 5. Studies included in the systematic review of the effect of biological drugs on health biomarkers in patients afflicted by diffuse cutaneous systemic sclerosis.
Table 5. Studies included in the systematic review of the effect of biological drugs on health biomarkers in patients afflicted by diffuse cutaneous systemic sclerosis.
First Author, Year of Publication, Country, and DrugStudy DesignParticipants (Baseline Sample Size, Age, Sex, Withdrawals, and Final Group Sample Size)InterventionOutcomesResults
Allanore et al., 2020 France [30]
Romilkimab
Phase II, randomized, double-blind, placebo-controlled clinical study20 ♂ and 77 ♀
>18 years
Study withdrawals: 10
no = 48 Romilkimab
no = 49 PBO
200 mg Romilkimab or PBO SC/week for 24 weeks.mRSS
FVC%
DLCO%
HAQ-DI
GI vs. GC:
↓* mRSS
↑ FVC %
↑ DLCO %
↓HAQ-DI score
Changes from baseline:
↓* mRSS
↓ FVC %
↓ DLCO %
↓HAQ-DI score
Gordon et al., 2018
USA [31]
Belimumab
Randomized, double-blind, placebo-controlled pilot studyn = 20
>18 years
Study withdrawals: 2
no = 7 Belimumab + MMF
no = 10 PBO + MMF
n = 6 Belimumab + MMF
n = 9 PBO + MMF
10 mg/kg Belimumab or PBO c/2 weeks first 3 doses and c/4 weeks until week 48 + 1000 mg 2 times/day MMF 48 weeksmRSS
FCV%
DLCO%
GI vs. GC:
↓ mRSS
↑ FVC %
↑ DLCO %
Changes from baseline:
↓* mRSS
↑* FVC %
↑* DLCO %
Khanna et al., 2018
USA [32]
Tocilizumab
Phase II, randomized, double-blind, placebo-controlled clinical trial20 ♂ and 67 ♀
>18 years
Study withdrawals: 36
no = 44 PBO
no = 43 TCZ
no = 31 PBO-TCZ
no = 30 TCZ-TCZ
n = 24 PBO-TCZ
n = 27 TCZ-TCZ
162 mg TCZ sc 48 weeks double blind + 162 mg TCZ sc 48 weeks open
period
total Mrss
HAQ-DI score
FCV%
DLCO %
GI vs. GC:
↓ total mRSS
↔ HAQ-DI score
↓ FVC%
↓ DLCO %
Changes from baseline:
↓ total mRSS
↓ HAQ-DI score
↓ FVC %
↓ DLCO %
Khanna et al., 2020
USA [33]
Abatacept
Phase II, randomized, double-blind, placebo-controlled clinical study22 ♂ and 66 ♀
>18 years
Study withdrawals: 12
no = 44 Abatacept
no = 44 PBO
n = 35 Abatacept
n = 34 PBO
125 mg Abatacept or PBO SC 1 times/week 12 monthsmRSS
FVC%
HAQ-DI
GI vs. GC:
↓ mRSS
↑ FVC %
↓HAQ-DI score
Changes from baseline:
↓ mRSS
↓ FVC %
↓ HAQ-DI score
Khanna et al., 2020
USA [34]
Riociguat
Phase II, randomized,
double-blind, placebo-controlled, international, multicenter clinical trial
29 ♂ and 92 ♀
>18 years
Study withdrawals: 33
no = 60 Riociguat
no = 61 PBO
n = 42 Riociguat
n = 46 PBO
0.5–2.5 mg Riociguat 3 times/day 52 weeksmRSS
FVC%
DLCO%
HAQ-DI
GI vs. GC:
↓ mRSS
↑ FVC %
↑ DLCO %
↓HAQ-DI score
Changes from baseline:
↓ mRSS
↓ FVC %
↓ DLCO %
↑ HAQ-DI score
Shima et al., 2018
Japan [35]
Tocilizumab
Randomized, open-label clinical trial10 ♂ and 3 ♀
20–65 years
Study withdrawals: 0
n = 7 TZC + conventional treatment
n = 6 conventional treatment
8 mg/kg/month TCZ 6 monthsDLCO %
mRSS
GI vs. GC:
↑DLCO %
↓ mRSS
Changes from baseline:
↓ DLCO %
↓ mRSS
Abbreviations: ↑ = no significant increase; ↓ = no significant decrease; ↔ = no significant change. ↑* = significant increase; ↓* = significant decrease; *: indicates significant values (p < 0.05); PBO: placebo; TCZ: tocilizumab; SC: subcutaneous; mRSS: modified Rodnan scale; HAQ-DI Score: Health Assessment Questionnaire; FVC: forced vital capacity; DLCO: diffusion test for carbon monoxide; MMF: micofenolato mofetil.
Table 6. Number of adverse effects of biological drugs in the intervention group.
Table 6. Number of adverse effects of biological drugs in the intervention group.
Adverse EffectsBiological Drugs
TocilizumabBelimumabRiociguatAbataceptRomilkimab
Infections 81
Cardiac1 1
Gastrointestinal1
Respiratory 1
Renal
Vascular1
Neoplasms 1
Musculoskeletal
Nervous
Endocrinologists
Psychiatric1
Reproductive
Blood and Lymphatics1
Dermatological
Deaths 1
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fernández-Lázaro, D.; Iglesias-Lázaro, M.; Garrosa, E.; Rodríguez-García, S.; Jerves Donoso, D.; Gutiérrez-Abejón, E.; Jorge-Finnigan, C. Impact of Innovative Treatment Using Biological Drugs for the Modulation of Diffuse Cutaneous Systemic Sclerosis: A Systematic Review. Medicina 2023, 59, 247. https://doi.org/10.3390/medicina59020247

AMA Style

Fernández-Lázaro D, Iglesias-Lázaro M, Garrosa E, Rodríguez-García S, Jerves Donoso D, Gutiérrez-Abejón E, Jorge-Finnigan C. Impact of Innovative Treatment Using Biological Drugs for the Modulation of Diffuse Cutaneous Systemic Sclerosis: A Systematic Review. Medicina. 2023; 59(2):247. https://doi.org/10.3390/medicina59020247

Chicago/Turabian Style

Fernández-Lázaro, Diego, María Iglesias-Lázaro, Evelina Garrosa, Saray Rodríguez-García, David Jerves Donoso, Eduardo Gutiérrez-Abejón, and Conrado Jorge-Finnigan. 2023. "Impact of Innovative Treatment Using Biological Drugs for the Modulation of Diffuse Cutaneous Systemic Sclerosis: A Systematic Review" Medicina 59, no. 2: 247. https://doi.org/10.3390/medicina59020247

Article Metrics

Back to TopTop