- Comment
- Open access
- Published:
CAR-T cell therapy for juvenile-onset autoimmune diseases: a promising future?
Arthritis Research & Therapy volume 27, Article number: 102 (2025)
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting B cells has shown promising results, including drug-free remission, in adult-onset autoimmune diseases. Extending this therapeutic approach to the pediatric population, particularly for juvenile autoimmune diseases, presents an exciting opportunity. However, challenges specific to juvenile-onset autoimmune conditions, such as long-term adverse events, heightened disease activity, and the imperative to reduce steroid exposure, must be considered. While this strategy appears viable for these severe conditions, the limited data available for this population and the absence of evidence on cases with a high genetic component, such as monogenic lupus, represent significant challenges. Most monogenic lupus cases are associated with innate immune defects, and the involvement of B cells in these genetic anomalies remains poorly understood. In this review, we examine the potential indications, current knowledge, and limitations of CAR-T cell therapy in juvenile-onset autoimmune diseases, extending the discussion beyond early-onset lupus.
Introduction
Early-onset autoimmune diseases exhibit distinct characteristics, often manifesting with greater severity compared to their adult-onset counterparts. Juvenile systemic lupus erythematosus (jSLE) typically presents with a more aggressive phenotype characterized by heightened disease activity and earlier onset of organ damage, with a frequent contribution of lupus nephritis and neurological involvement [1]. Severe forms of juvenile dermatomyositis (JDM) can evolve into chronically active diseases, with persistent disease activity extending years beyond the initial diagnosis. The identification of myositis-specific autoantibodies over the past decade has facilitated the classification of sub-phenotypes, each associated with specific clinical outcomes [2]. Patients with jSLE and JDM commonly experience a marked decline in quality of life [3, 4]. The management of autoimmune disease with juvenile-onset is particularly challenging, often requiring the use of combination therapies and prolonged corticosteroid. This approach can lead to significant immunosuppression, life-threatening complications, and an increased risk of infections and organ damage, all contributing to long-term disability [5,6,7].
While treatment strategies are frequently extrapolated from guidelines for adult-onset SLE and dermatomyositis, the management of these juvenile conditions requires specialized expertise tailored to the unique needs of younger patients. Achieving optimal disease control critical to support normal development, facilitate uninterrupted schooling and minimize long-term disease sequelae [8].
B cells play a pivotal role in the pathophysiology of SLE by presenting autoantigens to T cells and secreting pro-inflammatory cytokines [9] while plasmablasts contribute by generating autoantibodies. Immune complex deposits are critical in the development of renal pathology. Similarly, B lymphocytes are central in dermatomyositis evidenced by the significant reduction in regulatory B cells and the expansion of CD19 + B lymphocytes, which are activated via type-I interferons (IFN-I). These abnormalities in B cell compartments are closely correlated with clinical disease activity in JDM [10,11,12].
Targeting B lymphocytes with anti-CD20 therapies such as Rituximab, Obinutuzumab, and Ocrelizumab, has yielded variable results in SLE and JDM [13,14,15]. The limited efficacy observed may be due to the predominance of pathogenic B cells being plasmablasts, which express CD19 but not CD20, and the incomplete depletion of B cells in tissues achieved by monoclonal antibodies, particularly with rituximab [16].
Chimeric antigen receptor (CAR) T-cells represent an innovative approach in cellular therapy wherein T-cells are genetically engineered to express a chimeric antigen receptor that targets a specific membrane antigen. Specifically, anti-CD19 CAR T-cells are activated upon recognition of their target antigen, leading to their proliferation and potent cytotoxic action against CD19 + B cells. This cytotoxicity is mediated through various mechanisms, including perforin/granzyme and cytokine-induced cytotoxicity [17].
CAR-T cells targeting CD19 have been developed for the treatment of refractory or relapsed B cell malignancies in pediatric patients, with remarkable success [18]. Recently, anti-CD19 CAR-T cells have been used to treat adult patients with refractory auto-immune diseases such as SLE and DM [19]. This therapy can induce long-term drug-free remission, eliminate autoantibodies, and reset the B cell repertoire [19,20,21].
In early 2024, the first case reports emerged of children with refractory jSLE and JDM achieving similar remarkable results, including sustained remission following a few months of follow-up and without significant adverse effects [22,23,24].
The aim of this review is to provide a comprehensive overview of the use of CAR-T cells therapy for juvenile autoimmune diseases, to discuss current and future limitations, and to advocate for the advancement of these innovative approaches in pediatric rheumatology.
Specificities of the pediatric-onset autoimmune diseases
Juvenile systemic lupus erythematosus
JSLE is defined as lupus with onset before 18 years of age. The most common jSLE manifestations include hematological, cutaneous, musculoskeletal and renal symptoms [25,26,27]. Additionally, severe pulmonary, and life-threatening neuropsychiatric manifestations are also reported in jSLE [28, 29]. These patients are particularly susceptible to mental health issues, displaying a higher prevalence of fatigue, pain, anxiety, and depressive symptoms compared to the general population [30]. Compared to adults, jSLE is more frequently associated with hematological and renal complications; thrombocytopenia, hemolytic anemia, and lupus nephritis which are significantly more prevalent in this population [26, 31]. A large meta-analysis found that anti-dsDNA antibodies were more common in jSLE and disease activity, as measured by the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI), was higher in children than in adults at disease onset [32].
Clinical complications often develop within two years of diagnosis, and drug-free remission is rarely achieved in these patients. As they transition into adulthood, individuals with jSLE frequently experience impaired health-related quality of life, significantly impacted by changes in physical appearance. By their 20 s, approximately half of young adults with jSLE develop significant organ damage, including cerebrovascular accidents, renal transplants, replacement arthropathies, and myocardial infarctions (6). Standardized mortality rates among SLE patients, particularly those under 18, are higher than those of the general population, with mortality risk increasing with the duration of the disease [27, 33]. In the juvenile population, severe infections are the most common cause of death, whereas adults with SLE often succumb to cardiovascular events, renal failure, infections, or cancers [27, 34,35,36]. Furthermore, jSLE is more frequently associated to single gene mutations, defining the field of Mendelian lupus [37]. At least 7% of jSLE are monogenic, predominantly involving innate immune anomalies linked to complement defects or interferon dysregulation [38].
Various combination therapies have been recommended for jSLE, with new therapeutics recently entering the market. The Pediatric Rheumatology European Society (PreS) international task force now recommends the application of a'treat-to-target'strategy in jSLE to ensure long-term survival, prevent organ damage, and optimize health-related quality of life [39]. The treatment target should be disease remission, with low disease activity being acceptable in refractory cases, achieved with the lowest possible dosage of glucocorticoids.
The cornerstone of maintenance therapy in jSLE remains hydroxychloroquine often combined with Disease-Modifying Antirheumatic Drugs (DMARDs) such as methotrexate, mycophenolate mofetil and azathioprine despite the challenge of maintaining daily medication adherence in this population [40]. Severe forms of lupus (class III/IV or refractory lupus nephritis, neurolupus or pulmonary involvement) may require cyclophosphamide treatment, raising concerns about reproductive toxicity in pediatric patients [41,42,43,44,45,46]. Additionally, the long-term use of immunosuppressive treatments increases the risk of non-melanoma skin cancers and lymphomas [47, 48].
Therapeutic options for jSLE are limited, with only one biologic treatment (belimumab) approved, while rituximab is used off-label for refractory cases. Janus Kinase (JAK) inhibitors should be considered in cases of monogenic lupus, depending on the presence of specific mutations [39, 49,50,51,52] (Table 1). Novel therapeutics, such as obinutuzumab, voclosporine [53] and anifrolumab [54]. Have recently emerged for adult-onset SLE, although these medications are not yet approved for patients under 18 years of age. Despite advancements in treatment, a large number of children treated for jSLE continue to present with severe and refractory forms of the disease. No immunosuppressive treatment has been able to achieve both clinical and biological remission, which is why autologous hematopoietic stem cell transplantation (HSCT) is now being considered for refractory SLE [55].
Autologous HSCT, aimed at achieving broad immune depletion through conditioning treatments (e.g., anti-thymocyte globulin and cyclophosphamide or alemtuzumab), has been used in the treatment of SLE for the past 20 years. Between 1994 and 2018, 107 cases were reported in the European Society for Blood and Marrow Transplantation (EBMT) registry, including 18 patients under 18 years of age at the time of transplantation [56]. Auto-HSCT has been shown to enable the discontinuation of immunosuppressive treatment and achieve both clinical and serological remission. However, it is associated with significant risks and potential complications, including secondary autoimmune disorders and gonadotoxicity, with an estimated mortality rate of around 5% [57, 58]. Therefore, auto-HSCT is indicated only in the most severe and life-threatening cases. Allogeneic HSCT has been proposed for five patients (two children and three adults) with successful outcomes [59].
Juvenile dermatomyositis
Juvenile dermatomyositis (JDM) is the most prevalent idiopathic inflammatory myopathy in childhood, characterized by involvement of both muscle and skin with significant heterogeneity in clinical presentation and disease progression. JDM can manifest as monophasic, polycyclic, or persistently active disease [60], with severe cases leading to profound muscular deficits, respiratory dysfunction and swallowing disorders. associated with JDM can also cause gastrointestinal dysfunction, potentially escalating to life-threatening complications. A subgroup of patients with MDA5-positive antibody JDM and rapidly progressive interstitial lung disease (RP-ILD) may have a particularly severe prognosis [61]. The overall mortality rate is estimated at 3%, with variation among subgroups depending on autoantibodies [62].
The juvenile form is characterized by more pronounced vasculopathy, calcinosis, telangiectasias, and ulcerative skin disease at presentation compared to the adult form. Additionally, younger age at onset is associated with a trend toward more intensive treatment approaches in younger children with JDM [63].
Data from a large multicenter study conducted by UK specialists on over 500 patients with JDM suggests that the majority of patients (approximately 89%) show an overall improvement in disease activity over time. However, a smaller subgroup (around 11%) presents with more persistent disease activity lasting up to 10 years post-diagnosis [64]. Baseline factors associated with this more severe disease trajectory include higher levels of baseline disease activity, abnormal respiration, lipodystrophy, and a longer time from symptom onset to diagnosis. Muscle weakness, despite treatment and remission of other symptoms, is a common issue, leading to functional impairments in muscle strength and physical activity levels. These physical limitations can impact daily activities and overall quality of life [65].
The primary treatment for JDM relies on corticosteroid, often combined with immunosuppressants. Methotrexate is commonly used in conjunction with steroids (Table 2). As second-line treatment option, other DMARDs such as mycophenolate mofetil (MMF) and cyclosporine may be considered. In severe cases, intravenous immunoglobulins may be employed, particularly for managing swallowing difficulties [66]. The efficacy of Rituximab has been investigated in juvenile dermatomyositis, and its use is endorsed by experts [14, 67, 68].
Observations from retrospective case series have underscored the effectiveness of JAK inhibitors, such as baricitinib, which enable the tapering or discontinuation of steroids and other immunosuppressive agents [69,70,71]. Abatacept has shown improvement in disease activity in an open-label study [72]. Although rare, cases of autologous hematopoietic stem cell transplantation have suggested efficacy, though the data remain limited compared to those for jSLE [73].
In summary, jSLE and JDM are two pediatric diseases with significant unmet therapeutic needs. From a pathogenesis standpoint, B cells are considered a promising therapeutic target; however, the results of B-cell depletion using monoclonal antibodies have been inconsistent. This highlights the urgent need for the development of novel anti-B-cell therapies.
CAR T cells as an anti-B cell therapy
Chimeric antigen receptors are human-made receptors that combine two essential elements of the adaptive immune response: an antibody fragment combined with motifs involved in intracellular signaling so that induces a TCR-like antigen-specific and costimulation response [74, 75]. CARs consist of several parts. The first is the single-chain variable fragment (ScFv), which fuses the variable regions of heavy (VH) and light (VL) immunoglobulin chains that provide antigen specificity and are connected by a linker peptide. The ScFv is then linked to a hinge region (typically from CD8a or CD28) and a transmembrane domain (also CD8a or CD28). The presence or absence of additional co-stimulatory molecules (CD28 or 4 - 1BB) upstream of the CD3z chain distinguishes CAR generations [76]. These domains are absent in first-generation CARs, whereas second- and third-generation CARs possess one or two co-stimulatory domains, respectively [76]. To date, six CAR T-cell products have been authorized in France and Europe for use in hematologic B cells malignancies [77]. All commercial CAR-T-cells in the European Union are autologous and second-generation cells. The production process of autologous CAR-T cells is lengthy and complex [78]. The first step involved isolating the patient's leukocytes via apheresis. After transfer to production units, lymphocytes are sorted magnetically, activated with beads coated with anti-CD3 and CD28 antibodies and cytokines, and exposed to a viral vector that delivers the genetic sequence necessary for CAR expression in T cells. While various techniques exist for integrating Deoxyribonucleic acid (DNA) sequences into T cells [79], the most widely used for CAR-T generation employs lentiviral particles, which are known for their high transduction efficiency and low toxicity. After activation and transduction, the cells are maintained in culture for the expansion phase and then prepared for infusion. The main regulatory release controls are viability, CAR expression measurement, potency tests against antigen-expressing cells, and vector copy number (VCN) determination.
As previously mentioned, the recognition of a target expressing the antigen recognized by CAR mimics TCR-mediated activation [80]. This activation leads to the proliferation of CAR-T-cells and the secretion of cytokines (IL- 2, IFN-γ, and TNF-α), increasing their number and cytolytic effector functions [81]. The main pathway of cytotoxicity is the perforin/granzyme pathway [82]. In tumor models, it has also been shown that CAR-T-induced apoptosis can involve interactions between death receptors and their ligands (TRAIL, Fas-FasL) [83].
Targeting CD19 + B cells with CAR T cells in juvenile-onset autoimmune disease: evidence to date
Clinical presentation of adolescents treated with anti-CD19 CAR T cells therapy
On July 2024, it was reported that 11 patients with SLE had received treatment with anti-CD19 CAR T cells and one had received CD19-BCMA compound CAR. Of these patients, 10 were over 18 years old and 2 were under 18. All patients had severe and refractory disease, characterized by skin involvement and proliferative kidney nephritis (WHO grade III and IV). Notably, most of the adult patients experienced their first manifestations of the disease before the age of 18 (5 out 10 of adult patients) [19, 22, 23, 84,85,86]. Recently, a satisfactory response to anti-CD19 CAR T-cell therapy has been recently reported in four adults with inflammatory myositis, which included myositis and interstitial lung disease, with a maximum follow-up period of 12 months [19, 21].
Two adolescents with refractory jSLE have been treated with CAR T cell infusions. The first case, involving a 15-year-old girl with refractory jSLE, was reported in April 2024 by the Erlangen team [22]. A second patient was reported by the medical team at Bambino Gesu Children’s Hospital in Rome, Italy [23]. Additionally, a 12-year-old Caucasian boy with severe chronic JDM was successfully treated with CAR T cells [24]. The characteristics of all patients are detailed in Table 3.
The median age at the administration of CAR-T cell infusion was 15 years [range 12–15]. All patients exhibited multiple clinical manifestations including severe renal and/or pulmonary complications in SLE and severe skin and muscular involvement in cases of JDM.
Patients with jSLE had extremely elevated activity scores, with a median SLEDAI of 22.5 (range 22–23), which exceeded the median SLEDAI of 16 (range 8–16) observed in the adult cohort treated with CAR-T cells [21, 87]. These patients were refractory to both conventional and combination therapeutic regimens. All patients had previously undergone B-cell-targeted therapy with rituximab or belimumab, which yielded limited efficacy. Additionally, cyclophosphamide was administered to patients 2 and 3, but it failed to achieve clinical remission.
Treatments modalities
The washout period, defined as the discontinuation of immunosuppressive drugs, for juvenile patients ranged from 3 to 7 days prior to leukapheresis, accompanied by a reduction in corticosteroid dosage. This washout period aimed to optimize T cell harvesting and subsequent ex vivo expansion. Its duration was necessarily limited due to the severe and life-threatening nature of the patients'disease manifestations in the absence of treatment. Notably, the washout period was shorter than that used in the adult population (median of 3 weeks) but did not adversely affect the expansion or efficacy of the CAR-T cells. All patients’autologous T cells were transduced with a second-generation lentiviral anti-CD19 CAR vector and infused at a uniform dosage of 1 × 106 CAR cells/kg, consistent with the protocol for patients with B cell malignancies and most reported autoimmune patients [19]. The lymphodepletion regimen varied, with differences in the duration and intensity of cyclophosphamide and fludarabine administration (Table 4). A"higher intensity"cyclophosphamide regimen was preferred in patients without renal failure. Despite the relatively short washout period, the procedure was successfully completed.
B Cell depletion and reconstitution
Peripheral B cell aplasia was observed between days 5 and 7 in all patients. Bone marrow aspirates at weeks 2 and 4 confirmed complete tissular B-cell depletion in patients 2 and 3. Patient 1 experienced persistent B cell aplasia for a period of 6 months, whereas patient 3 exhibited detectable B cells 8 weeks post-infusion. These B cells exhibited a phenotype of naïve mature cells with low levels of memory B cells, plasmablasts and mature B cells by week 18. Throughout the follow-up period, Patient 3’s IgG and IgM levels remained within normal limits. Unfortunately, no information regarding B-cell reconstitution was available for patient 2.
Clinical improvement
Patient 1, who had severe renal disease, was able to discontinue dialysis on day 17, and exhibited a significant improvement of estimated glomerular filtration rate (eGFR) (only mild persistent proteinuria, resolution of oedema, significant decrease of proteinuria). Additionally, the patient experienced normalized appetite, gained muscle mass, and saw a complete resolution of arthritis, with the SLEDAI score reaching 0 a few weeks after CAR-T cell infusion. Patient 2 showed normalization of cardiac function and renal markers, with the SLEDAI score decreasing to 2 after 6 weeks of treatment. The patient with dermatomyositis initially experienced a worsening of skin and muscular symptoms due to the tapering of immunosuppressants. However, by week 4 post-infusion, the disease activity score began to decrease, eventually leading to the normalization of muscle strength and resolution of cutaneous manifestations, including calcinosis and skin ulcerations. By the last follow-up, all patients had successfully discontinued all immunosuppressive therapies.
Biological assessment
All patients (n = 3) exhibited normalization of laboratory parameters, including the dramatic decrease or disappearance of anti-double-stranded DNA antibodies. The IFN score normalization in the patient with dermatomyositis was observed as early as week 24 and was sustained thereafter. As mentioned above, CK levels were already at the baseline before the CAR-T cell infusion and did not increase after discontinuation of corticosteroids/immunosuppressive drug.
Side effects
All juvenile patients (n = 3) experienced similar side effects, including neutropenia (n = 1 grade 3, n = 2 grade 4), transient anemia (n = 3 grade 2), and mild cytokine release syndrome (CRS, n = 3 grade 1). One patient required a single infusion of Tocilizumab (8 mg/kg) for grade 1 CRS management. None of the patients exhibited signs of infections or immune effector cell-associated neurotoxicity syndrome (ICANS) (Fig. 1).
Illustration of the treatment pathway for patients with severe early-onset autoimmune diseases that are refractory to conventional treatments, with CAR-T cell therapy serving as a salvage option. To optimize CAR-T cell expansion, lymphodepletion is induced using fludarabine and cyclophosphamide which carries a risk of disease exacerbation. Following CAR-T cell infusion, patients commonly experience a transient period of bone marrow aplasia, increasing susceptibility to infections. The expansion of CAR-T cells leads to the targeted depletion of autoreactive B lymphocytes. However, this process may also trigger the release of cytokines, potentially leading to cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Lymphocyte reconstitution occurs progressively over the course of several weeks, though the timeline may vary among patients. The reconstituted B cells are naive and no longer produce autoreactive antibodies, leading to both clinical and biological remission, thereby eliminating the need for ongoing immunosuppressive therapy. Figure created with Biorender.com
Challenges in pediatric CAR-T cell use
Children who received CAR-T cell therapy had particularly severe and refractory autoimmune diseases, unresponsive to conventional therapies. These patients also suffered severe complications related to intensive immunosuppressive therapies and high-dose corticosteroids, including arterial hypertension, osteoporosis, and infections. Prior treatment with the B-cell-depleting anti-CD20 monoclonal antibody Rituximab provided only partial and transient benefits. However, anti-CD19 CAR T-cell therapies appeared to be more effective (albeit with short follow-up), achieving B-cell clearance in the bone marrow. Notably, a dramatic response was observed in cases of severe renal disease in SLE and calcinosis in JDM, highlighting the therapeutic potential of CAR-T cells in these challenging complications.
Despite the promising outcomes, the limited number of treated patients and the short duration of follow-up necessitate confirmation through larger cohorts and randomized studies.
Conventional treatments have failed to achieve persistent normalization of biological markers such as anti-dsDNA antibodies and the type I interferon signature. The significant reduction in the IFN-I signature following CAR-T cell therapy suggests that IFN-I signaling may be a consequence, rather than a cause, of autoreactive B cell activation.
The hypothesis that CAR-T cell therapy resets the B-cell compartment, potentially eliminating autoreactive pathogenic B-cell clones, may explain the observed dramatic response observed. Post-treatment, a virtual disappearance of IgG and IgA expressing B cells are observed while IgM and IgD expressing B cell clone are expanded [88]. Interestingly, preexisting humoral immunity to vaccine related antigens can persist in adult patients suggesting a persistence of long-lived plasma cells and humoral immunity [89]. Additionally, repertoire analysis showed that CD19 CAR-T cell therapy did not affect the T cell receptor (TCR) repertoire allowing a normal T cell response [88].
The observed efficacy in a young patient with seronegative juvenile dermatomyositis, who had no detectable autoantibodies, suggests a role for B-cells or the presence of undetectable autoantibodies in this patient.
Leukapheresis can be challenging due to high disease activity and severe lymphopenia resulting from both the disease and prior immunosuppressive treatments. It is noteworthy that the washout period was shorter in juvenile patients compared to the adult population without compromising the efficacy of CAR-T cell therapy. Determining the optimal duration of the washout period will be critical for future pediatric studies.
While lymphodepletion with fludarabine and cyclophosphamide may have contributed to initial outcomes, the doses were lower than those used in previous treatments (e.g., plasmapheresis, cyclophosphamide), which had proven ineffective.
Given the heightened risk of infections, side effects, and treatment burdens, exploring reduced conditioning regimens is worthwhile. However, lymphodepletion remains necessary to induce CAR-T cell expansion. Although reducing the intensity of lymphodepletion may lessen side effects, such as ICANS, it could also reduce efficacy. Lymphodepletion might be particularly beneficial in conditions like myositis and scleroderma, where T-cell activation during CAR-T cell expansion could temporarily worsen disease activity.
Conditioning regimens were adjusted according to renal function, indicating a tailored approach to patient safety. Recent data regarding ovarian dysfunction with intravenous cyclophosphamide used for 6 months at the NIH regimen (0.5–0.75 g/m2 monthly) minimally impacts fertility, suggesting that fertility concerns related to CAR-T cell therapy, which involves even lower doses of cyclophosphamide, are likely minimal [90].
The persistence of CAR-T cells for six months in the bloodstream of the pediatric patient from Erlangen, is notably longer than observed in adults. However, in autoimmune diseases, prolonged CAR-T cell persistence or extended B-cell aplasia may not be necessary, as immune system resetting appears to induce prolonged remission in adults effectively. The reconstitution of the naive B-cell population aims to restore humoral immunity and enable vaccine protection. Notably, remission persisted in the juvenile dermatomyositis patient even after B-cell reconstitution at eight months, without the need for ongoing immunosuppression.
No deaths were reported in either the adult or juvenile cohorts during CAR-T cell therapy. The survival rate appears notably higher than with autologous or allogeneic HSCT for autoimmune diseases, although further studies are needed to confirm this, given the limited number of patients treated with CAR-T cells. Additionally, no severe side effects were reported, even in patients with end-stage kidney failure. In the juvenile population, mild cytokine release syndrome (CRS) occurred but resolved quickly with a single dose of Tocilizumab in one patient. Adults with anti-synthetase syndrome experienced some ICANS, but symptoms were effectively managed with a short course of dexamethasone [20]. The use of corticosteroids or Tocilizumab does not seem to affect the efficacy of CAR-T cell therapy in the treatment of hematological conditions [91].
CRS is a systemic condition characterized by a high cytokines production that can mimic infectious conditions, leading to fever, tachycardia, tachypnea, and hypotension. Following lymphodepletion, which induces profound neutropenia and B-cell depletion, along with the use of steroids or biologics (such as Tocilizumab), patients are at an increased risk of infectious complications [92]. The duration of B-cell aplasia varies and can last for several months. No infections have been reported in the juvenile population, but preventive measures, such as immunoglobulin replacement therapy starting one month after infusion, are recommended to avoid complications. Vaccine guidelines for children post-CAR-T cell therapy established in oncological indications, advise against administering live or attenuated vaccines during B-cell depletion due to reactivation risks, while inactivated vaccines should generally be postponed during B-cell aplasia. However, the influenza vaccine can be administered one month after CAR-T cell therapy [92].
Concerns have been raised regarding the safety of CAR-T cell therapies, particularly in relation to secondary malignancies and potential impacts on future pregnancies. The hypothesis that genetic modifications of T lymphocytes could induce T-cell neoplasms emerged following a Food and Drug Administration (FDA) warning about secondary T-cell malignancies in patients with hematologic disorders treated with CAR-T cells [93]. However, it is important to note that the adult population had significant prior exposure to chemotherapeutic agents, and pre-cancerous cells with clonal hematopoiesis were detected before CAR-T cell infusion, a condition likely less prevalent in the pediatric auto-immune population. Additionally, T-cell malignancies did not consistently express the CAR transgene [94]. A global survey on the risk of secondary malignancies after CAR-T cells therapy in pediatric oncology published reassuring data with only one patient identified to develop T cell lymphoma without retroviral transgene in the clonal population. No insertional mutagenesis was reported in pediatric patients [95] though further long-term studies are necessary to fully assess the enduring safety profile of CAR-T cell therapies.
The sustained drug-free remission observed after a single infusion strongly suggests a causal effect of CAR T-cell therapy, positioning the pediatric population as a promising candidate for future studies. The therapy has demonstrated a favorable safety profile and efficacy, with manageable side effects. Further research involving larger patient cohorts will be necessary to confirm these findings and refine treatment protocols.
Multidisciplinary teams, including pediatric rheumatologists and hematologists, are crucial for managing these severe conditions with multi-organ involvement and tailoring treatments to meet the specific needs of each patient.
To our knowledge, none of the patients treated with CD19-targeted therapy currently described have undergone genetic exploration. The early onset of the disease and its severity suggest that genetic factors might be implicated. JSLE cases have been linked to single gene mutations, defining the concept of monogenic or Mendelian lupus. Genes associated with monogenic lupus can be classified into at least five functional categories: complement deficiency, interferonopathies driven by defective nucleic acid metabolism or innate sensor overactivity, tolerance breakdown of the adaptive immune system, JAK-STAT pathway abnormalities, apoptosis dysregulation, and TLR-mediated autoimmunity [96, 97].
Notable differences can be observed between monogenic and polygenic lupus. Apart from complement deficiencies, certain mutations in lupus patients lead to immune deficiencies such as IKAROS, RAG2, while others are commonly associated with lymphoproliferative diseases like PRKCδ. PKCδ deficiency is the first identified B cell–related form of monogenic lupus [98]. Some mutations, such as those in C1q, C1R, C1S, PRKCD, UNC93B1, and TLR7, are more frequently associated with glomerulonephritis [96, 97, 99]. In monogenic lupus associated with B cells genetic defects, the strategy might be harmful, although the risk of relapse is likely higher than in adult-onset non-genetic lupus. Some monogenic interferonopathies have been described with an increase in lupus-like phenotype as a direct consequence of excessive production of IFN-I [100]. The penetrance is variable, suggesting that constitutive expression of IFN-I alone is not sufficient to drive the autoimmunity. Consequently, resetting B cells in these patients may still represent an option to cure a flare and delay disease progression.
The duration of remission achieved through CAR-T cell therapy in patients with monogenic lupus remains unclear. In the worst-case scenario, genetic factors may facilitate early relapses. However, it is also recognized that additional external environmental factors likely contribute to the onset of clinical manifestations. Given the severity of these clinical manifestations, achieving multiple years of remission without any treatment would be considered a significant success. In the event of a relapse, a second infusion could be a viable option.
In March 2024, the first CAR T-cell trial in children with lupus gets FDA go-ahead. This marks the first CAR T-cell trial for pediatric lupus in the United States, with Seattle Children's planning to enroll patients in the summer of 2024 [101].
Simultaneously, a prospective open-label single-arm study is underway in China to evaluate the efficacy of anti-CD19 CAR-T cells in children aged 5 to 18 years with refractory SLE (NTC06222853). This trial, spanning 24 months, consists of two phases for dose exploration and expansion to determine a safe and effective dose, with an expected enrollment of 10 to 19 patients. Further clinical trials are essential to develop a treatment algorithm that enables the timely and optimal utilization of CAR T-cell therapies.
CAR-T cell therapies have been used in a limited number of pediatric patients with SLE and JDM, selected based on disease activity and severity. Early-onset SLE is often complicated by neurolupus, while JDM is associated with interstitial lung disease and gastrointestinal vasculopathy. To date, CAR-T cells have not been applied to these specific complications. Further exploration of CAR-T cell therapy in these indications and in other autoimmune diseases, such as systemic scleroderma, is warranted.
CAR-T cell therapies offer significant advantages over conventional treatments, offering impressive clinical outcomes with drug-free remission, and potentially reducing long-term health complications and the economic burden associated with managing chronic conditions in patients with early-onset disease. Treatment adherence, a significant challenge in adolescents, is effectively addressed with CAR-T therapies. The ability to return to school within a few months after infusion is another significant benefit. However, the high cost of manufacturing CAR-T cells restricts their widespread use. Therefore, conducting economic studies valuating the long-term benefits of clinical remission for severe patients are essential.
Although short-term safety data are encouraging, long-term safety evaluations remain crucial. New strategies utilizing allogeneic CAR (T or NK) cells could enhance the accessibility of such therapies. Additionally, non-cellular therapies, such as bispecific T cell engagers (BiTEs), are currently being investigated in adult autoimmunity and may potentially be adapted for pediatric use.
Conclusions
CAR-T cells represent a significant breakthrough in pediatric rheumatology, offering the potential for curative treatments for autoimmune diseases. This new era in patient care has brought forth novel questions and challenges, particularly regarding outcomes in patients with specific genetic backgrounds. Coordinated translational and clinical research is essential to deepen our understanding and expand access to CAR-T cell therapies for children with severe and refractory conditions. Understanding and expand access to CAR-T cell therapies for children with severe and refractory conditions. While most of the efforts currently focus on adult-onset autoimmune diseases, the high burden and severity in children make this population of particular interest. Consequently, there is an urgent need to offer CAR-T cell therapy to severe pediatric patients, ideally through clinical trials, or the least, through compassionate use.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- BiTEs:
-
Bispecific T cell Engagers
- CAR:
-
Chimeric Antigen Receptor
- CAT:
-
Cutaneous Assessment Tool
- CAT -BM:
-
Cutaneous Assessment Tool in the shortened Binary Method version
- CK:
-
Creatine Kinase
- CMAS:
-
Childhood Myositis Assessment Scale
- CRS:
-
Cytokine Release Syndrome
- CYC:
-
Cyclophosphamide
- DMARD:
-
Disease-Modifying Antirheumatic Drugs
- DNA:
-
Deoxyribonucleic Acid
- EBMT:
-
European Society for Blood and Marrow Transplantation
- eGFR:
-
Estimated Glomerular Filtration Rate
- FDA:
-
Food and Drug Administration
- GC:
-
Glucocorticoids
- HCQ:
-
Hydroxychloroquine
- HSCT:
-
Hematopoietic Stem Cell Transplantation
- ICANS:
-
Immune Effector Cell-Associated Neurotoxicity Syndrome
- IFN-I:
-
Type-I Interferons
- JAK:
-
Janus Kinase
- JDM:
-
Juvenile Dermatomyositis
- jSLE:
-
Juvenile Systemic Lupus Erythematosus
- MMF:
-
Mycophenolate Mofetil
- MTX:
-
Methotrexate
- NIH:
-
National Institutes of Health
- PRES:
-
Pediatric Rheumatology European Society
- RP-ILD:
-
Rapidly progressive interstitial lung disease
- ScFv:
-
Single-chain variable Fragment
- SLEDAI:
-
Systemic Lupus Erythematosus Disease Activity
- SHARE:
-
Single Hub and Access point for pediatric Rheumatology in Europe.
- TCR:
-
T Cell Receptor
- VCN:
-
Vector Copy Number
- VH:
-
Variable regions of Heavy immunoglobulin
- VL:
-
Variable regions of Light immunoglobulin
References
Smith EMD, Lythgoe H, Midgley A, Beresford MW, Hedrich CM. Juvenile-onset systemic lupus erythematosus: update on clinical presentation, pathophysiology and treatment options. Clin Immunol. 2019;209:108274. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clim.2019.108274.
Papadopoulou C, Chew C, Wilkinson MGL, McCann L, Wedderburn LR. Juvenile idiopathic inflammatory myositis: an update on pathophysiology and clinical care. Nat Rev Rheumatol. 2023;19:343–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41584-023-00967-9.
Ruperto N, Buratti S, Duarte-Salazar C, Pistorio A, Reiff A, Bernstein B, et al. Health-related quality of life in juvenile-onset systemic lupus erythematosus and its relationship to disease activity and damage. Arthritis Care Res. 2004;51:458–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.20412.
Witczak BN, Bollerslev J, Godang K, Schwartz T, Flatø B, Molberg Ø, et al. Body composition in long-standing juvenile dermatomyositis: associations with disease activity, muscle strength and cardiometabolic measures. Rheumatology. 2022;61:2959–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keab805.
Smith EMD, Egbivwie N, Jorgensen AL, Ciurtin C, Al-Abadi E, Armon K, et al. Real world treatment of juvenile-onset systemic lupus erythematosus: Data from the UK JSLE cohort study. Clin Immunol. 2022;239:109028. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clim.2022.109028.
Costa-Reis P, Nativ S, Isgro J, Rodrigues T, Yildirim-Toruner C, Starr A, et al. Major infections in a cohort of 120 patients with juvenile-onset systemic lupus erythematosus. Clin Immunol. 2013;149:442–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clim.2013.08.009.
Knight AM, Trupin L, Katz P, Yelin E, Lawson EF. Depression risk in young adults with juvenile- and adult-onset lupus: twelve years of followup. Arthritis Care Res. 2018;70:475–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/acr.23290.
Beresford MW, Davidson JE. Adolescent development and SLE. Best Pract Res Clin Rheumatol. 2006;20:353–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.berh.2005.11.003.
Wilkinson MGL, Rosser EC. B cells as a therapeutic target in paediatric rheumatic disease. Front Immunol. 2019;10:214. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2019.00214.
Li W, Tian X, Lu X, Peng Q, Shu X, Yang H, et al. Significant decrease in peripheral regulatory B cells is an immunopathogenic feature of dermatomyositis. Sci Rep. 2016;6:27479. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep27479.
Piper CJM, Wilkinson MGL, Deakin CT, Otto GW, Dowle S, Duurland CL, et al. CD19+CD24hiCD38hi B cells are expanded in juvenile dermatomyositis and exhibit a pro-inflammatory phenotype after activation through toll-like receptor 7 and interferon-α. Front Immunol. 2018;9:1372. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.01372.
Syntakas AEK, Nguyen HD, Restuadi R, Jebson BR, Alexiou V, Marshall LR, et al. OA34 Unravelling the role of B cells and interferon dysregulation in juvenile dermatomyositis. Rheumatology. 2024;63:keae163-034. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keae163.034.
Furie RA, Aroca G, Cascino MD, Garg JP, Rovin BH, Alvarez A, et al. B-cell depletion with obinutuzumab for the treatment of proliferative lupus nephritis: a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis. 2022;81:100–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/annrheumdis-2021-220920.
Aggarwal R, Bandos A, Reed AM, Ascherman DP, Barohn RJ, Feldman BM, et al. Predictors of clinical improvement in rituximab-treated refractory adult and juvenile dermatomyositis and adult polymyositis. Arthritis Rheumatol. 2014;66:740–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.38270.
Mysler EF, Spindler AJ, Guzman R, Bijl M, Jayne D, Furie RA, et al. Efficacy and safety of ocrelizumab in active proliferative lupus nephritis: results from a randomized, double-blind, Phase III Study. Arthritis Rheum. 2013;65:2368–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.38037.
Mei HE, Schmidt S, Dörner T. Rationale of anti-CD19 immunotherapy: an option to target autoreactive plasma cells in autoimmunity. Arthritis Res Ther. 2012;14:S1. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/ar3909.
Montalvo MJ, Bandey IN, Rezvan A, Wu K-L, Saeedi A, Kulkarni R, et al. Decoding the mechanisms of chimeric antigen receptor (CAR) T cell-mediated killing of tumors: insights from granzyme and Fas inhibition. Cell Death Dis. 2024;15:1–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-024-06461-8.
Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1709866.
Müller F, Taubmann J, Bucci L, Wilhelm A, Bergmann C, Völkl S, et al. CD19 CAR T-cell therapy in autoimmune disease — a case series with follow-up. N Engl J Med. 2024;390:687–700. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa2308917.
Taubmann J, Knitza J, Müller F, Völkl S, Aigner M, Kleyer A, et al. Rescue therapy of antisynthetase syndrome with CD19-targeted CAR-T cells after failure of several B-cell depleting antibodies. Rheumatology. 2024;63:e12–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/kead330.
Müller F, Boeltz S, Knitza J, Aigner M, Völkl S, Kharboutli S, et al. CD19-targeted CAR T cells in refractory antisynthetase syndrome. Lancet. 2023;401:815–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(23)00023-5.
Krickau T, Naumann-Bartsch N, Aigner M, Kharboutli S, Kretschmann S, Spoerl S, et al. CAR T-cell therapy rescues adolescent with rapidly progressive lupus nephritis from haemodialysis. Lancet. 2024;403:1627–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(24)00424-0.
Marasco E, Bracaglia C, Merli P, Alvarez PM, Nicolai R, Algeri M, et al. P101 Anti-CD19 CAR-T cell therapy for refractory childhood-onset systemic lupus erythematosus. Poster Present., Lupus Foundation of America; 2024, p. A113.1-A113. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/lupus-2024-el.155.
Nicolai R, Merli P, Moran Alvarez P, Bracaglia C, Del Bufalo F, Marasco E, et al. Autologous CD19‐ targeting CAR T cells in refractory juvenile dermatomyositis. Arthritis Rheumatol 2024:art.42933. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.42933.
Tucker LB, Menon S, Schaller JG, Isenberg DA. Adult- and childhood-onset systemic lupus erythematosus: a comparison of onset, clinical features, serology, and outcome. Rheumatology. 1995;34:866–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/34.9.866.
Bader-Meunier B, Armengaud JB, Haddad E, Salomon R, Deschênes G, Koné-Paut I, et al. Initial presentation of childhood-onset systemic lupus erythematosus: a French multicenter study. J Pediatr. 2005;146:648–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jpeds.2004.12.045.
Charras A. Systemic lupus erythematosus in children and young people. Curr Rheumatol Rep. 2021;23:1–5.
Palafox-Flores JG. Systemic lupus erythematosus in pediatric patients: Pulmonary manifestations. Respir Med. 2023;220:107456.
Labouret M, Costi S, Bondet V, Trebossen V, Le Roux E, Ntorkou A, et al. Juvenile neuropsychiatric systemic lupus erythematosus: identification of novel central neuroinflammation biomarkers. J Clin Immunol. 2023;43:615–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10875-022-01407-1.
Quilter M, Hiraki L, Korczak D. Depressive and anxiety symptom prevalence in childhood-onset systemic lupus erythematosus: a systematic review. Lupus. 2019;28:878–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0961203319853621.
Ambrose N, Morgan TA, Galloway J, Ionnoau Y, Beresford MW, Isenberg DA. Differences in disease phenotype and severity in SLE across age groups. Lupus. 2016;25:1542–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0961203316644333.
Livingston B, Bonner A, Pope J. Differences in autoantibody profiles and disease activity and damage scores between childhood- and adult-onset systemic lupus erythematosus: a meta-analysis. Semin Arthritis Rheum. 2012;42:271–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semarthrit.2012.05.001.
Watson L, Leone V, Pilkington C, Tullus K, Rangaraj S, McDonagh JE, et al. Disease activity, severity, and damage in the UK juvenile-onset systemic lupus erythematosus cohort. Arthritis Rheum. 2012;64:2356–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.34410.
Amaral B, Murphy G, Ioannou Y, Isenberg DA. A comparison of the outcome of adolescent and adult-onset systemic lupus erythematosus. Rheumatology. 2014;53:1130–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/ket488.
Chiche L, Jourde-Chiche N, Bader-Meunier B, Retornaz F, Malaekah S, Eb M, et al. Acute pancreatitis as a cause of mortality in pediatric systemic lupus erythematosus: results of a multiple cause-of-death analysis in France. Semin Arthritis Rheum. 2016;46:e6-7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semarthrit.2016.03.009.
Klein A, Cimaz R, Quartier P, Decramer S, Niaudet P, Baudouin V, et al. Causes of death in pediatric systemic lupus erythematosus. Clin Exp Rheumatol. 2009;27:538–9.
Tusseau M, Khaldi-Plassart S, Cognard J, Viel S, Khoryati L, Benezech S, et al. Mendelian causes of autoimmunity: the lupus phenotype. J Clin Immunol. 2024;44:99. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10875-024-01696-8.
Belot A, Rice GI, Omarjee SO, Rouchon Q, Smith EMD, Moreews M, et al. Contribution of rare and predicted pathogenic gene variants to childhood-onset lupus: a large, genetic panel analysis of British and French cohorts. Lancet Rheumatol. 2020;2:e99-109. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S2665-9913(19)30142-0.
Smith EMD, Aggarwal A, Ainsworth J, Al-Abadi E, Avcin T, Bortey L, et al. Towards development of treat to target (T2T) in childhood-onset systemic lupus erythematosus: PReS-endorsed overarching principles and points-to-consider from an international task force. Ann Rheum Dis. 2023;82:788–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/ard-2022-223328.
Semo-Oz R, Wagner-Weiner L, Edens C, Zic C, One K, Saad N, et al. Adherence to medication by adolescents and young adults with childhood-onset systemic lupus erythematosus. Lupus. 2022;31:1508–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/09612033221115974.
Groot N, De Graeff N, Marks SD, Brogan P, Avcin T, Bader-Meunier B, et al. European evidence-based recommendations for the diagnosis and treatment of childhood-onset lupus nephritis: the SHARE initiative. Ann Rheum Dis. 2017;76:1965–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/annrheumdis-2017-211898.
Fanouriakis A, Pamfil C, Sidiropoulos P, Damian L, Flestea A, Gusetu G, et al. Cyclophosphamide in combination with glucocorticoids for severe neuropsychiatric systemic lupus erythematosus: a retrospective, observational two-centre study. Lupus. 2016;25:627–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0961203315622821.
Arıcı ZS, Batu ED, Ozen S. Reviewing the recommendations for lupus in children. Curr Rheumatol Rep. 2015;17:17. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11926-014-0489-5.
Shin JI, Lee KH, Park S, Yang JW, Kim HJ, Song K, et al. Systemic lupus erythematosus and lung involvement: a comprehensive review. J Clin Med. 2022;11:6714. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm11226714.
Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, Garrido EDR, Danieli MG, et al. Immunosuppressive therapy in lupus nephritis: the euro-lupus nephritis trial, a randomized trial of low-dose versus high-dose intravenous cyclophosphamide. Arthritis Rheum. 2002;46:2121–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.10461.
Austin HA, Klippel JH, Balow JE, Le Riche NGH, Steinberg AD, Plotz PH, et al. Therapy of lupus nephritis. N Engl J Med. 1986;314:614–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJM198603063141004.
Van Den Reek JMPA, Van Lümig PPM, Janssen M, Schers HJ, Hendriks JCM, Van De Kerkhof PCM, et al. Increased incidence of squamous cell carcinoma of the skin after long-term treatment with azathioprine in patients with auto-immune inflammatory rheumatic diseases. J Eur Acad Dermatol Venereol. 2014;28:27–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jdv.12041.
Bernatsky S, Ramsey-Goldman R, Joseph L, Boivin J-F, Costenbader KH, Urowitz MB, et al. Lymphoma risk in systemic lupus: effects of disease activity versus treatment. Ann Rheum Dis. 2014;73:138–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/annrheumdis-2012-202099.
Tambralli A, Beukelman T, Cron RQ, Stoll ML. Safety and efficacy of rituximab in childhood-onset systemic lupus erythematosus and other rheumatic diseases. J Rheumatol. 2015;42:541–6. https://doiorg.publicaciones.saludcastillayleon.es/10.3899/jrheum.140863.
Hogan J, Godron A, Baudouin V, Kwon T, Harambat J, Deschênes G, et al. Combination therapy of rituximab and mycophenolate mofetil in childhood lupus nephritis. Pediatr Nephrol. 2018;33:111–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00467-017-3767-4.
Guzman M, Hui-Yuen JS. Management of pediatric systemic lupus erythematosus: focus on belimumab. Drug Des Devel Ther. 2020;14:2503–13. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/DDDT.S216193.
Thuner J, Cognard J, Belot A. How to treat monogenic SLE? Best Pract Res Clin Rheumatol. 2024;38:101962. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.berh.2024.101962.
Rovin BH, Teng YKO, Ginzler EM, Arriens C, Caster DJ, Romero-Diaz J, et al. Efficacy and safety of voclosporin versus placebo for lupus nephritis (AURORA 1): a double-blind, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet. 2021;397:2070–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(21)00578-X.
Furie RA, Morand EF, Bruce IN, Manzi S, Kalunian KC, Vital EM, et al. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): a randomised, controlled, phase 3 trial. Lancet Rheumatol. 2019;1:e208–19. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S2665-9913(19)30076-1.
Stojkic I, Harper L, Coss S, Kallash M, Driest K, Lamb M, et al. CAR T cell therapy for refractory pediatric systemic lupus erythematosus: a new era of hope? Pediatr Rheumatol. 2024;22:1–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12969-024-00990-4.
Jessop H, Farge D, Saccardi R, Alexander T, Rovira M, Sharrack B, et al. General information for patients and carers considering haematopoietic stem cell transplantation (HSCT) for severe autoimmune diseases (ADs): a position statement from the EBMT Autoimmune Diseases Working Party (ADWP), the EBMT Nurses Group, the EBMT Patient, Family and Donor Committee and the Joint Accreditation Committee of ISCT and EBMT (JACIE). Bone Marrow Transplant. 2019;54:933–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41409-019-0430-7.
Doglio M, Alexander T, Del Papa N, Snowden JA, Greco R. New insights in systemic lupus erythematosus: from regulatory T cells to CAR-T-cell strategies. J Allergy Clin Immunol. 2022;150:1289–301. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2022.08.003.
Daikeler T, Tichelli A, Passweg J. Complications of autologous hematopoietic stem cell transplantation for patients with autoimmune diseases. Pediatr Res. 2012;71:439–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/pr.2011.57.
Greco R, Labopin M, Badoglio M, Veys P, Furtado Silva JM, Abinun M, et al. Allogeneic HSCT for autoimmune diseases: a retrospective study from the EBMT ADWP, IEWP, and PDWP Working Parties. Front Immunol. 2019;10:1570. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2019.01570.
Bader-Meunier B, Gitiaux C, Belot A, Brochard K, Mouy R, Ponce D, et al. French expert opinion for the management of juvenile dermatomyositis. Arch Pédiatrie. 2019;26:120–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.arcped.2018.12.002.
Nishi K, Ogura M, Tamai N, Gima M, Ide K, Koinuma G, et al. Successful rituximab treatment for severe rapidly progressive interstitial lung disease with anti-MDA5 antibody-positive juvenile dermatomyositis: a case report and literature review. Pediatr Rheumatol. 2022;20:60. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12969-022-00723-5.
Ravelli A, Trail L, Ferrari C, Ruperto N, Pistorio A, Pilkington C, et al. Long-term outcome and prognostic factors of juvenile dermatomyositis: a multinational, multicenter study of 490 patients. Arthritis Care Res. 2010;62:63–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/acr.20015.
Martin N, Krol P, Smith S, Beard L, Pilkington CA, Davidson J, et al. Comparison of children with onset of juvenile dermatomyositis symptoms before or after their fifth birthday in a UK and Ireland juvenile dermatomyositis cohort study. Arthritis Care Res. 2012;64:1665–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/acr.21753.
Deakin CT, Papadopoulou C, McCann LJ, Martin N, Al-Obaidi M, Compeyrot-Lacassagne S, et al. Identification and prediction of novel classes of long-term disease trajectories for patients with juvenile dermatomyositis using growth mixture models. Rheumatology. 2021;60:1891–901. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keaa497.
Boros C, McCann L, Simou S, Cancemi D, Ambrose N, Pilkington CA, et al. Juvenile dermatomyositis: what comes next? Long-term outcomes in childhood myositis from a patient perspective. Pediatr Rheumatol. 2022;20:102. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12969-022-00754-y.
Enders FB, Bader-Meunier B, Baildam E, Constantin T, Dolezalova P, Feldman BM, et al. Consensus-based recommendations for the management of juvenile dermatomyositis. Ann Rheum Dis. 2017;76:329–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/annrheumdis-2016-209247.
Aggarwal R, Loganathan P, Koontz D, Qi Z, Reed AM, Oddis CV. Cutaneous improvement in refractory adult and juvenile dermatomyositis after treatment with rituximab. Rheumatology. 2017;56:247–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/kew396.
Pediatric Rheumatologist Collaborators, Spencer C, Rouster-Stevens K, Gewanter H, Syverson G, Modica R, et al. Biologic therapies for refractory juvenile dermatomyositis: five years of experience of the Childhood Arthritis and Rheumatology Research Alliance in North America. Pediatr Rheumatol. 2017;15:50. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12969-017-0174-0.
Le Voyer T, Gitiaux C, Authier F-J, Bodemer C, Melki I, Quartier P, et al. JAK inhibitors are effective in a subset of patients with juvenile dermatomyositis: a monocentric retrospective study. Rheumatology. 2021;60:5801–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keab116.
Wang Z, Zheng Q, Xuan W, Xu X, Lu M, Wu J, et al. Short-term effectiveness of baricitinib in children with refractory and/or severe juvenile dermatomyositis. Front Pediatr. 2022;10:962585. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fped.2022.962585.
Solignac M, Cabrera N, Fouillet-Desjonqueres M, Duquesne A, Laurent A, Foray A-P, et al. JAK inhibitors in refractory juvenile rheumatic diseases: efficacy, tolerance and type-I interferon profiling, a single center retrospective study. J Autoimmun. 2024;147:103248. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaut.2024.103248.
Curiel RV, Nguyen W, Mamyrova G, Jones D, Ehrlich A, Brindle KA, et al. Improvement in disease activity in refractory juvenile dermatomyositis following abatacept therapy. Arthritis Rheumatol. 2023;75:1229–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.42450.
Delemarre EM, van den Broek T, Mijnheer G, Meerding J, Wehrens EJ, Olek S, et al. Autologous stem cell transplantation aids autoimmune patients by functional renewal and TCR diversification of regulatory T cells. Blood. 2016;127:91–101. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2015-06-649145.
Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86:10024–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.86.24.10024.
Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90:720–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.90.2.720.
Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3:388–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2159-8290.CD-12-0548.
Baguet C, Larghero J, Mebarki M. Early predictive factors of failure in autologous CAR T-cell manufacturing and/or efficacy in hematologic malignancies. Blood Adv. 2024;8:337–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/bloodadvances.2023011992.
Wang X, Rivière I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics. 2016;3:16015. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/mto.2016.15.
Atsavapranee ES, Billingsley MM, Mitchell MJ. Delivery technologies for T cell gene editing: applications in cancer immunotherapy. EBioMedicine. 2021;67:103354. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebiom.2021.103354.
Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol. 2002;20:70–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt0102-70.
Korell F, Berger TR, Maus MV. Understanding CAR T cell-tumor interactions: paving the way for successful clinical outcomes. Med N Y N. 2022;3:538–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.medj.2022.05.001.
Davenport AJ, Jenkins MR, Cross RS, Yong CS, Prince HM, Ritchie DS, et al. CAR-T cells inflict sequential killing of multiple tumor target cells. Cancer Immunol Res. 2015;3:483–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2326-6066.CIR-15-0048.
Mamonkin M, Rouce RH, Tashiro H, Brenner MK. A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies. Blood. 2015;126:983–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2015-02-629527.
Taubmann J, Müller F, Yalcin Mutlu M, Völkl S, Aigner M, Bozec A, et al. CD19 chimeric antigen receptor T cell treatment: unraveling the role of b cells in systemic lupus erythematosus. Arthritis Rheumatol. 2024;76:497–504. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.42784.
Zhang W, Feng J, Cinquina A, Wang Q, Xu H, Zhang Q, et al. Treatment of systemic lupus erythematosus using BCMA-CD19 compound CAR. Stem Cell Rev Rep. 2021;17:2120–3. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12015-021-10251-6.
Mougiakakos D, Krönke G, Völkl S, Kretschmann S, Aigner M, Kharboutli S, et al. CD19-targeted CAR T cells in refractory systemic lupus erythematosus. N Engl J Med. 2021;385:567–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMc2107725.
Boulougoura A, Gendelman H, Surmachevska N, Kyttaris VC. Journal Club: Anti-CD19 chimeric antigen receptor T cell therapy for refractory systemic lupus erythematosus. ACR Open Rheumatol. 2023;5:624–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/acr2.11614.
Wilhelm A, Chambers D, Müller F, Bozec A, Grieshaber-Bouyer R, Winkler T, et al. Selective CAR-T cell mediated B cell depletion suppresses interferon signature in SLE. JCI Insight. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci.insight.179433.
Bhoj VG, Arhontoulis D, Wertheim G, Capobianchi J, Callahan CA, Ellebrecht CT, et al. Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood. 2016;128:360–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2016-01-694356.
Sharma SK, Jain S, Bahl P, Potturi P, Rathi M, Naidu S, et al. Ovarian dysfunction with moderate-dose intravenous cyclophosphamide (modified NIH regimen) and mycophenolate mofetil in young adults with severe lupus: a prospective cohort study. Arthritis Res Ther. 2020;22:189. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-020-02292-y.
Costa BA, Flynn J, Nishimura N, Devlin SM, Farzana T, Rajeeve S, et al. Prognostic impact of corticosteroid and tocilizumab use following chimeric antigen receptor T-cell therapy for multiple myeloma. Blood Cancer J. 2024;14:84. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41408-024-01048-0.
Los-Arcos I, Iacoboni G, Aguilar-Guisado M, Alsina-Manrique L, Díaz De Heredia C, Fortuny-Guasch C, et al. Recommendations for screening, monitoring, prevention, and prophylaxis of infections in adult and pediatric patients receiving CAR T-cell therapy: a position paper. Infection. 2021;49:215–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s15010-020-01521-5.
Willyard C. Do cutting-edge CAR-T-cell therapies cause cancer? What the data say Nature. 2024;629:22–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/d41586-024-01215-0.
Hamilton MP, Sugio T, Noordenbos T, Shi S, Bulterys PL, Liu CL, et al. Risk of second tumors and T-cell lymphoma after CAR T-Cell therapy. N Engl J Med. 2024;390:2047–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa2401361.
Lamble AJ, Schultz LM, Nguyen K, Hsieh EM, McNerney K, Rouce RH, et al. Risk of T-cell malignancy after CAR T-cell therapy in children, adolescents, and young adults. Blood Adv. 2024;8:3544–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/bloodadvances.2024013243.
Omarjee O, Picard C, Frachette C, Moreews M, Rieux-Laucat F, Soulas-Sprauel P, et al. Monogenic lupus: dissecting heterogeneity. Autoimmun Rev. 2019;18:102361. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.autrev.2019.102361.
Wolf C, Lim EL, Mokhtari M, Kind B, Odainic A, Lara-Villacanas E, et al. UNC93B1 variants underlie TLR7-dependent autoimmunity. Sci Immunol. 2024;9:eadi9769. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciimmunol.adi9769.
Belot A, Kasher PR, Trotter EW, Foray A-P, Debaud A-L, Rice GI, et al. Protein kinase cδ deficiency causes mendelian systemic lupus erythematosus with B cell-defective apoptosis and hyperproliferation. Arthritis Rheum. 2013;65:2161–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.38008.
David C, Badonyi M, Kechiche R, Insalaco A, Zecca M, De Benedetti F, et al. Interface gain-of-function mutations in TLR7 cause systemic and neuro-inflammatory disease. J Clin Immunol. 2024;44:60. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10875-024-01660-6.
Crow YJ, Manel N. Aicardi-Goutières syndrome and the type I interferonopathies. Nat Rev Immunol. 2015;15:429–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri3850.
Burki T. CAR T-cell therapy for SLE in children. Lancet Rheumatol. 2024:S2665991324000985. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S2665-9913(24)00098-5.
Acknowledgements
The authors thank Samira Plassart for her contribution to Figure 1.
This article was written for the'Club des Immunothérapies Innovantes dans les IMIDS'(C3I) consortium.
The C3I Consortium is composed of the following members:
Gaetane NOCTURNE: gaetane.nocturne@aphp.fr
David LAUNAY: david.launay@chru-lille.fr
Benjamin TERRIER: benjamin.terrier@aphp.fr
Camille BIGENWALD: camille.bigenwald@gustaveroussy.fr
Christophe RICHEZ: christophe.richez@chu-bordeaux.fr
Noémie JOURDE-CHICHE: noemie.JOURDE@ap-hm.fr
Sylvain CHOQUET: sylvain.choquet@aphp.fr
Divi CORNEC: divi.cornec@chu-brest.fr
Jean SIBILIA: jean.sibilia@chru-strasbourg.fr
Sylvain CHOQUET: sylvain.choquet@aphp.fr
Thierry MARTIN: thierry.Martin@chru-strasbourg.fr
Samuel BITOUN: samuel.bitoun@aphp.fr
Zahir AMOURA: zahir.amoura@aphp.fr
Olivier HERMINE: olivier.hermine@aphp.fr
Xavier MARIETTE: xavier.mariette@aphp.fr
Emmanuel DONNADIEU: emmanuel.donnadieu@inserm.fr
Philippe BOUSSO: philippe.bousso@pasteur.fr
Grégory PUGNET: pugnet.g@chu-toulouse.fr
Justine DECROOCQ: justine.decroocq@aphp.fr
Edouard FORCADE: edouard.forcade@chu-bordeaux.fr
Alain MEYER: alain.meyer1@chru-strasbourg.fr
Christian JORGENSEN: christian.jorgensen@inserm.fr
Laurent PEYRIN-BIROULET: contact@laurent-peyrinbiroulet.com
Alexandre BELOT: alexandre.belot@chu-lyon.fr
Maurine JOURET: maurine.jouret@chu-lyon.fr
Jerome AVOUAC: jerome.avouac@aphp.fr
Marc SCHERLINGER: marc.scherlinger@chru-strasbourg.fr
Funding
This research was funded by ANR (ANR- 21-CE17 - 0064 [SOCSIMMUNITY]]), ANR- 21-RHUS- 08 [COVIFERON]) from the ANR–Recherche Hospitalo Universitaire Program; by the Horizon Europe (01057100 [UNDINE]) from the HORIZON-HLTH- 2021-DISEASE- 04; le Centre de référence des rhumatismes inflammatoires, des interféronopathies et des maladies autoimmunes (RAISE).
Author information
Authors and Affiliations
Consortia
Contributions
M.J. wrote the manuscript draft. A.B., S.V., B.F., S.B., J.A. and M.S. revised the manuscript. All authors contributed to the article and approved the submitted version.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
MJ is a consultant for Novartis. MS is a consultant for Abbvie, Amgen, AstraZeneca, Biogen, Bristol Myers Squibb, Fresenius Kabi, Galapagos, Innate Pharma, Nordic Pharma, Novartis, Sandoz. JA: Honoraria: Galapagos, Lilly, Pfizer, Abbvie, Bristol-Myers Squibb, Sanofi, Roche-Chugai, Nordic Pharma, Medac, Novartis, Biogen, Fresenius Kabi, Janssen, and MSD. Research grants: Bristol-Myers Squibb, Pfizer (Passerelle), Novartis (Dreamer), Fresenius Kabi, Galapagos, Nordic Pharma. AB is a consultant for Kabi, Roche Chugai, GlaxoSmithKline, AbbVie, Novartis, and Boehringer Ingelheim.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Jouret, M., Viel, S., Fournier, B. et al. CAR-T cell therapy for juvenile-onset autoimmune diseases: a promising future?. Arthritis Res Ther 27, 102 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-025-03564-1
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-025-03564-1