touchIMMUNOLOGY touchIMMUNOLOGY
Systemic Lupus Erythematosus
Read Time: 15 mins

CAR T Cell Therapy: A New Hope for Refractory Lupus?

Copy Link
Published Online: Sep 12th 2023 touchREVIEWS in RMD. 2024;3(1):13–20 DOI: http://doi.org/10.17925/RMD.2023.2.2.1
Authors: Michael Alsharkawy, Alice Mason, May Lwin, Christopher J Edwards
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Systemic lupus erythematosus (SLE) is a complex multisystem, autoimmune disorder. The disease involves multiple inflammatory and immune pathways, posing several treatment challenges. The role of B cells in the pathogenesis and disease activity of SLE is well described, and treatments targeting B cells are now well established. Despite advances in therapeutics for SLE, the clinical course of SLE can be associated with significant morbidity and mortality. For many individuals, the management of SLE requires lifelong immunosuppression. Chimeric antigen receptor (CAR) T cell therapy is an emerging treatment for haematological malignancies that uses CAR T cells to target CD19+ B lymphocytes. CAR T cell therapy has shown promising results in murine models of SLE, and a case report and subsequent case series in people with SLE reporting drug-free remission following CAR T cell therapy against CD19+ B cells have recently been published. This review discusses CAR T cell therapy, comparing it with established B cell depletion therapies, such as rituximab, and exploring its safety, limitations and promise in the management of patients with SLE.

Keywords

Autoimmune disordersB cell depletionchimeric antigen receptorschimeric antigen receptor T cell therapyfeasibilityimmunotherapyrefractory lupussystemic lupus erythematosus.

Article:

Systemic lupus erythematosus (SLE) is a multisystem, autoimmune disorder affecting skin, joints, kidneys, the heart, lungs, the brain and blood cells, and is characterized by heterogeneous presentations and severity. It is more common in females than males, with a ratio of 9:1, and it is associated with significant morbidity and mortality.1 Despite recent novel therapies being licensed for the management of SLE,2 there is still an unmet need for therapeutic agents with low toxicity that can control the disease and prevent recurrence, particularly for refractory cases.3

The therapeutic challenges relating to SLE can be attributed to the disease’s complex pathways and the interplay between the factors involved in its development.4 Genetics is perhaps the starting point in SLE development, with multiple modes of contribution. Although single gene defects can be implicated in SLE,5,6 they may not necessarily cause the disease on their own. Over 100 gene loci with polymorphisms have been identified7 and are thought to be associated with SLE development, antinuclear antibody and anti-double-stranded (ds) DNA formation, T cell activation and regulation, and interferon production.8–10

A major driver of SLE development is the loss of tolerance of various components of the adaptive immune system and the subsequent formation of B cell clones producing target autoantibodies, the most notable being anti-dsDNA antibodies in increased quantities.11,12 And a key step is believed to be the aberrant apoptosis, cell clearing and pathologic T helper and T cytotoxic lymphocyte stimulation, with reduced T regulatory function.13,14 Aberrant apoptosis exposes the immune system to various intranuclear components and gives rise to the various antinuclear antibodies commonly associated with SLE and SLE-like illnesses.15

Aside from genetic factors, hormones, smoking, ultraviolet exposure and viruses also contribute to the complex pathophysiology of SLE.16–20 For example, oestrogen upregulates T helper cytokines that activate B cells.21,22 It also acts on increasing expression of B cell activating factor (BAFF and Blys) and interferon signature genes, thus increasing inflammation. While viral antigens such as Epstein–Barr virus can mimic the molecular structure of common SLE antigens; patients with SLE have been shown to have a dysregulated immune response against Epstein–Barr virus.23

B cell depletion in systemic lupus erythematosus

Rituximab, a chimeric monoclonal antibody targeting CD20+ B lymphocytes, was approved in 1997 for the treatment of low-grade B cell non-Hodgkin’s lymphoma.24 The role played by B cells in the pathogenesis of SLE had been recognized by then.25,26 In individuals with SLE, B lymphocytes not only produce autoantibodies, they have also been shown to have various altered characteristics compared with the normal population.27 For example, B lymphocytes in individuals with SLE display an uneven increased amount of class‐switched memory B cells compared with naïve B cells. There is also an exaggerated B cell receptor response with a lower activation threshold, allowing autoreactive B cells to thrive with minimal antigen contact. As such, the potential role of B lymphocyte depletion therapy in autoimmune diseases such as SLE was considered.28 In 2002, Leandro et al.29 treated six female patients with active SLE resistant to standard immunosuppressive therapy on an open-label basis with two 500 mg infusions of rituximab, cyclophosphamide, and high-dose oral corticosteroids. The success of this approach supported the new understanding that, although T cytotoxic and T helper cells play a major role in the development and progression of the autoimmune process, B lymphocytes play an equally clinically significant role in the production of autoantibodies that perpetuate the autoimmune cascade.

The 2008 European Alliance of Associations for Rheumatology guidelines for the management of SLE recommended the use of rituximab for patients with lupus nephritis who are resistant to cyclophosphamide therapy.30 This recommendation is based on small, non-controlled trials with a short follow-up suggesting that up to 50% of patients with SLE refractory to cyclophosphamide for lupus nephritis may have a clinically significant response to rituximab.31,32 Over multiple iterations of subsequent society guidelines, B cell depletion therapy (such as rituximab) was established as a therapeutic option for patients with autoimmune diseases (including SLE), in particular in patients with SLE who did not respond to or could not tolerate other established therapies (such as cyclophosphamide or mycophenolate mofetil).33 This recommendation was also included in the latest guidelines published by the European Alliance of Associations for Rheumatology in 202034 and the British Society for Rheumatology in 2018.35

Expanding upon the role of B cells in SLE, new molecular targets such as B cell growth factors, including B-cell activating factor (also known as B-lymphocyte stimulator), were identified and targeted,36 leading to the development and licensing of belimumab for the treatment of SLE.

Other B cell-targeting therapies have been studied. These include the anti-CD19 monoclonal antibody, obexelimab,37 and the anti-CD22 monoclonal antibody, epratuzumab.38 Therapies targeting additional activation factors were also tried: a proliferating-inducing ligand, atacicept39; inhibitors of B cell intracellular activation pathways, (for example, pathways involving Bruton’s tyrosine kinase), such as fenebrutinib40; and B cell proteasome inhibitors, such as bortezomib.41 These treatments were unsuccessful due to either not meeting their endpoints or their safety profile and adverse events.42 The only new drug to be licensed for the treatment of SLE since belimumab has been anifrolumab, a human monoclonal antibody targeting type I interferon receptor subunit 1.43,44

Outcomes in patients with SLE remain poor, with significant mortality and morbidity.45–47 Some cases remain difficult to manage, and a significant portion of the patient population continues to exhibit variable treatment responses. Furthermore, those showing favourable initial outcomes almost always require on-going maintenance therapy after induction to prevent disease relapse. New therapies are needed for treating resistant SLE. This review explores an exciting emerging therapy for B cell depletion: chimeric antigen receptor (CAR) T cell therapy.

What is chimeric antigen receptor T cell therapy?

CAR T cell therapy is a treatment that has been established for the management of several haematological malignancies, including B cell lymphomas and multiple myeloma, with research looking at implementing this safely and effectively in solid organ tumours.48–51 The principle behind CAR T cell therapy is using the body’s own immune system, which in this case are the CD4+ T helper and CD8+ T cytotoxic lymphocytes, to perform specific programmable therapeutic functions.

The concept of CAR T cell therapy was first described in 1989 by Gross et al.52 CARs were an attempt to utilize and direct the specificity of T cells, in a non-major histocompatibility complex (MHC)-restricted manner. This process provides the T cell with an antibody-like specificity as well as the ability to transmit effectively the signal for T cell activation and execution of its effector functions. At the same time, it allows the T cells to bypass cellular processing and presentation, which would normally be done via antigen-presenting cells expressing MHC class II (in the case of CD4+ T cells) or MHC class I restriction (in the case of CD8+ T cells).

In its current form, CAR T cell therapy is a complex process. The principles and methods behind CAR T cell therapy administration vastly differ from conventional pharmacological therapeutics or receptor targeting therapies via monoclonals such as rituximab. This leads to a unique array of challenges, treatment side effects and potentially different long-term outcomes.

CAR T cell therapy begins with a process called leukapheresis, in which the patient’s white blood cells are harvested.48 The population of CD4+ and CD8+ T lymphocytes is then isolated from the harvested cells using cell processors or centrifugation-based cell separators. Then, a virus carrying genetic material with the instructions for producing CARs, such as a retrovirus or lentivirus, is introduced and transfects the extracted T lymphocytes. The viral genetic material carries the instruction for the formation and expression of the CARs on the surface of the T cells. These CARs that will be expressed on the surface of the T cells are not a naturally occurring receptor. They are a fusion protein of a selected single-chain fragment of the variable (binding) region from a specific monoclonal antibody and one or more T cell receptor intracellular signaling domains, hence the name “chimeric”, i.e., having a part of a B cell receptor/antibody domain in the extracellular part and then connected via a transmembrane domain to a T cell stimulator domain in the intracellular part (Figure 1).

Figure 1:The structure of a chimeric antigen receptor

Once the T cells have been transfected with the instructions for the CARs and the CARs have been expressed on the T cell surface, they undergo a process of stimulation and expansion to increase the number of T cells.53,54 Over 7–12 days, the cells are multiplied in number to achieve clinically significant amounts of CAR T cells. Among the approved CAR T cell therapies, tisagenlecleucel (KYMRIAH®; Novartis, Basel, Switzerland) can have a dose of between 1.2×106 to 6×108 cells per kg of body weight55 and ciltacabtagene autoleucel (CARVYKTI®; Janssen Biotech, Inc. Horsham, PA, USA) a dose of 0.5–1.0×106 cells per kg of body weight, with a maximum dose of 1×108 CAR-positive viable T cells per single infusion.56 After expansion, the CAR T cells undergo purification. The purification process removes other cells that are still in the preparation, as well as different proteins and substances that were used in the previous preparation steps. Finally, the CAR T cells are then re-infused back into the patient (Figure 2).

Figure 2:Steps involved in CAR T cell therapy preparation and administration

Pre-administration conditioning is a separate process that happens at the same time as steps 1–7.Steps 1–7 are the extraction and preparation of the cells and then the administration back into the patient. As this is happening they also get the pre-adminstration conditioning of cyclophosphamide and fludarabine.

CAR = chimeric antigen receptor; CD = cluster differentiation.

Prior to infusion, the patient must be conditioned and treated with lymphodepleting agents, such as cyclophosphamide or fludarabine. This aims to minimize the profound immunological response that occurs once the CAR T cells are infused, and the patient’s immune system is recruited by the CAR T cells against the tumour or the target antigen. The process from leukapheresis to the administration of the CAR T cells to the patient may take between 14 and 28 days.

The CAR T cell therapy products approved by the United States Food and Drug Administration57 and the European Medicines Agency58 as of March 2023 are detailed in Table 1.

Table 1:Therapies approved by the United States Food and Drug Administration and European Medicines Agency as of 13 March 2023

Year first approved by the FDA

Year first approved by the EMA

Brand name

Manufacturer

Generic name

Target antigen

Indication

2017

2018

KYMRIAH®

Novartis, Basel, Switzerland

tisagenlecleucel

CD19

B cell lymphomas

2017

2018

YESCARTA®

Kite Pharma Inc., Los Angeles, CA, USA

axicabtagene ciloleucel

CD19

B cell lymphomas

2020

2020

TECARTUS®

Kite Pharma Inc., Los Angeles, CA, USA

brexucabtagene autoleucel

CD19

B cell lymphomas

2021

2021

ABECMA®

Bristol Myers Squibb, New York, NY, USA

idecabtagene vicleucel

BCMA

Multiple myeloma

2021

2022

BREYANZI®

Juno Therapeutics, Inc., Seattle, WA, USA

lisocabtagene maraleucel

CD19

B cell lymphomas

2022

2022

CARVYKTI®

Janssen Biotech, Inc. Horsham, PA, USA

ciltacabtagene autoleucel

BCMA

Multiple myeloma

BCMA = B-cell maturation antigen ;CD = cluster differentiation;EMA = European Medicines Agency;FDA = Food and Drug Administration.

In the United Kingdom, the National Health Service and the National Institute for Health and Care Excellence currently recommend axicabtagene, tisagenlecleucel59 and brexucabtagene autoleucel60 for the treatment of B cell lymphomas.

Chimeric antigen receptor T cell therapy, a new concept in B cell depletion in the treatment of systemic lupus erythematosus

The potential for applying CAR T cell therapy to the treatment of SLE was first recognized in 2019 by Kansal et al.,61 who showed its feasibility and efficacy in mice models. They demonstrated that, by injecting CAR T cells to target CD19+ B cells in mice with SLE, the mice were able to live to the age of 18 months. This outcome differed significantly from that of mice that did not receive the treatment and had to be euthanized for advanced disease. This study also showed improvements in proteinuria, splenomegaly and reduction in anti-dsDNA antibody levels. These findings were supported by a further murine model62 and by the case of a 41-year-old female with a 20-year history of SLE who developed stage IV diffuse large B cell lymphoma and was treated with CAR T cell therapy against CD19.63 Following treatment, the woman from the case study achieved remission, which was confirmed by bone marrow aspirate and flow cytometry. The patient’s B cells remained at undetectable levels until Day 198, with recovery to normal levels at 9 months post therapy. Despite discontinuing prednisone and immunosuppression, the patient’s C3 and C4 complement levels remained within normal limits. Various antinuclear antibody titres were highly elevated prior to therapy; however, nuclear, cytoplasmic and granular antinuclear antibodies were undetectable for up to 37 weeks. The remission of both the lymphoma and SLE was seen up to 23 months following the administration of the CAR T cell therapy.

In 2021, the New England Journal of Medicine published a case report of a patient with SLE treated with CAR T cell therapy.64 The patient had World Health Organization class IIIA lupus nephritis (i.e. focal proliferative disease with active lesions), pericarditis, pleurisy, rash, arthritis and a history of Libman–Sacks endocarditis. The patient had previously been treated with hydroxychloroquine, high-dose glucocorticoids, cyclophosphamide, mycophenolate mofetil, tacrolimus, and the B cell-targeting therapies belimumab and rituximab. Despite these treatments, the disease was not controlled; consequently, the patient was treated with CAR T cell therapy targeting CD19+ B cells. Within 5 weeks of receiving the infusion, the patient achieved clinical and biochemical remission. At 44 days post infusion, the patient remained in remission.

In November 2022, Nature Medicine published a report by Mackensen et al.,65 which detailed the treatment of five cases of refractory SLE with CAR T cell therapy. The report describes how these patients had failed multiple prior treatments: pulsed glucocorticoids (5/5), hydroxychloroquine (5/5), mycophenolate mofetil (5/5), belimumab (5/5), cyclophosphamide (3/5) and azathioprine (2/5). Only one of the patients had previously received rituximab.

Both studies reported clinical improvement along with normalization of complement levels, the disappearance of anti-dsDNA antibodies and the resolution of proteinuria. Both studies also found that all patients achieved remission with CAR T cell therapy without needing further immunosuppressive therapy after several months of follow-up. In the five patient case series, the drug-free remission continued in the follow-up period, despite B cell reconstitution, which occurred on average after 110 ± 32 days. The follow-up periods in the case series were 17, 12, 8, 7 and 5 months.

These studies are the first therapeutic attempts at applying CAR T cell therapy in SLE in humans and provide an impressive and remarkable result. The treatment was offered under compassionate use, and it could be postulated that the patients would have experienced significant morbidity and perhaps even mortality without this intervention.

CAR T cell therapy advantages over established therapies

In the context of SLE treatment, CAR T cell therapy is in essence a B cell depleting therapy similar to rituximab. In reality, there are several stark differences.

The first difference is due to the very nature of rituximab being a chimeric protein and its impact on its binding to CD20 receptors.66 The other is the reduced ability of rituximab to target tissue B cells.67 This means that some B cells evade depletion when CD20 is targeted with rituximab.

Thirdly, rituximab targets CD20, while CAR T cell therapy targets CD19. Although many B cells express both antigens, mature B cells and plasma cells start to lose CD20 expression but continue to express CD19 while migrating into tissues.68 This suggests that CAR T cell therapy targeting CD19+ B cells offers a more significant depletion of B cells and plasma cells and thus a more complete ‘immune reset’.69,70

Finally, rituximab’s half-life is 22 days and is subject to various pharmacokinetic and pharmacodynamic effects that can limit its response and, hence, whether it has a lasting effect.71 Patients may also develop antidrug antibodies against rituximab, limiting repeated use.72 CAR T cells, on the other hand, are living cells that go on to exert a more lasting effect.73 Their persistence and the factors dictating their long-term behaviour can be influenced via alterations to the design of the CARs. For example, including CD28 in the CAR structure appears to generate effector memory cells. Newer CAR designs are created to improve efficacy and limit toxicity while striving to achieve tumour-specific functions.74 How all these advances in the CAR design will directly affect the treatment of patients with SLE remains to be seen.

Side effects and safety

CAR T cell therapy is characterized by a profound immune response led by the T cells. This immune response has been associated with well-described syndromes, namely cytokine release syndrome (CRS), neurotoxicity, cytopenias and hypogammaglobulinaemia.75–79

The flurry of immune cell activation that occurs when the CAR T cells are introduced leads to an abundance of activated immune cells and the overproduction of pro-inflammatory cytokines. This causes symptoms ranging from hypotension and increased transaminases to vascular leak syndrome, acute respiratory distress syndrome and encephalopathy. Patients with CRS were found to have very high levels of interleukin (IL)-6, leading to the approval of tocilizumab for the treatment of post-CAR T infusion CRS.80 CRS is now graded according to severity, with identifiable risk factors predicting the onset of CRS and its severity.81

The latest grading system proposed by the American Society for Transplantation and Cellular Therapy is based on the presence of fever, hypotension and hypoxia.82 Severity ranges from grade 1 to grade 4 depending on fever, the number of vasopressors needed and the amount of oxygen supplementation therapy required.

Neurotoxicity may also occur following CRS, with leakage of various immune cell components crossing the blood–brain barrier.78,79 Patients with neurotoxicity typically experience headaches, confusion, aphasia, dysphasia or somnolence. In addition to IL-6 antagonists, IL-1 antagonists were also found to be effective in the treatment of neurotoxicity.

Overall, nearly 100% of patients receiving CAR T cell therapy against CD19+ B lymphocytes to treat B cell lymphoma develop CRS, with at least 10% developing grade 3 to grade 4 CRS.83 Despite this, a Cochrane Review of 13 studies with a total of 679 participants receiving CAR T cell therapy for relapsed or refractory diffuse large B cell lymphoma found that no deaths were reported due to treatment.84 In the six patients with SLE treated with CAR T cell therapy targeting CD19+ B cells, side effects related to CAR T cell therapy were minimal. Five patients with SLE had no or only mild CRS. Fever (grade 1 CRS) occurred in only three of the six patients, and one patient only required a single infusion of 8 mg per kg body weight tocilizumab, with immediate cessation of the symptoms. None of the patients developed neurotoxicity. An explanation offered by Mackensen et al.65 as to why the patients with SLE receiving CAR T cell therapy had lesser side effects is the lower B cell burden in patients with SLE compared with patients with active B cell malignancies.

CAR T cell therapy targeted against CD19 causes the depletion of CD19+ B lymphocytes. This depletion can lead to hypogammaglobulinaemia and cytopenia.85 There is currently no evidence on how to manage this specifically. Prophylactic immunoglobulins are not currently recommended. Vaccinations are indicated before administering CAR T cell therapy targeting CD19, as with existing B cell-depleting treatments.

During the coronavirus disease 2019 pandemic, patients that received CAR T cell therapy were tested for antibody spike titres against the severe acute respiratory syndrome coronavirus 2. In one study, only 29% of patients were able to mount a clinically relevant antibody response.86 Clinically relevant vaccine responses appeared to improve with repeated vaccine doses.87

Other conditions that have been linked to B cell depletion therapies, such as progressive multifocal leukoencephalopathy, were found to occur in 0.9 cases per 1,000 recipients of CAR T cell therapy targeting CD19.88 This is much less common in patients receiving rituximab.89

Although the six patients with SLE treated with CAR T cell therapy have only experienced relatively minimal side effects to date, larger trials with longer follow-up periods are needed.

Practicality and feasibility

Although CAR T cell therapy usage is increasing, it is associated with several limitations and barriers, especially within the context of severe refractory autoimmune disorders.90,91

First, to administer CAR T cell therapy, the T cells must initially be extracted from the patient to undergo leukapheresis, transfection, expansion and then re-administration into the patient’s bloodstream (Figure 2). The administration itself must be preceded by leuko-ablation or conditioning to prevent CRS and immunotoxicity. The administration period, which is usually up to 14 days, may be too long for some patients with SLE who may require more urgent management.

Second, CAR T cell therapy is dependent on extracting T cells in specific quantities and of a certain quality and purity. However, patients with active SLE may experience alterations to their immune homeostasis or co-existing infections that prevent this. Nonetheless, this risk appears to be theoretical, and recent evidence shows that successful extraction is possible and feasible.92

Finally, the cost may also be a limit to the widespread implementation of CAR T cell therapy in its current form. Currently, the cost of each treatment exceeds £280,000.93 The cost of CAR T cell therapy may potentially become more affordable over the coming years; however, the current cost represents a barrier to accessing this treatment for patients with SLE, not only for compassionate use indications but also for running robust trials to compare it with existing therapies.

The future of CAR T cell therapy

CAR T cell therapy has undergone several improvements in both design and implementation.94 There are now several generations of CAR T cell receptors with various receptor designs and co-stimulatory domains.

Two of the limitations discussed in the previous section, namely the delay in achieving and administering the final product and ensuring the successful extraction of the patient’s T cells, could be overcome by using non-autologous CAR T cells, otherwise known as universal CAR (UCAR) T cells.95 UCAR T cells can be readily administered on demand via a ready-to-use T cell product carrying specific CAR molecules. Using these T cells, therefore, would enable the timely administration of CAR T cell therapy for urgent cases. However, UCAR T cells are not yet available for use in CAR T cell therapy and will need to be designed to overcome both graft versus host disease and graft rejection, among other challenges.

Furthermore, CAR T cell receptors are now being engineered to express different CAR receptors with multiple targets. Alternatively, the CAR T cell product may contain different T cell populations, each with a different CAR receptor.96 This can be applied in oncology for overcoming tumour evasion. However, it remains unclear how this can be applied in autoimmune disorders such as SLE.

Moving away from T cells, research is also underway on using other types of cells, such as CAR natural killer (NK) therapy or even macrophages.97 These alternative therapies would use NK cells or macrophages with engineered CAR receptors to target specific tumour antigens or, potentially, immune receptors associated with autoimmune diseases. NK cells and macrophages exhibit innate antitumour activity and do not induce graft versus host disease. NK cells also have wider immune-activating pathways and cytotoxicity mechanisms, including for solid organs.

Perhaps one of the more ambitious advances in CAR T cell therapy is in vivo CAR generation.98 Rather than extracting the T cells, which involves transfecting and expanding the T cells in vitro and then re-introducing them back into the patient, in vivo CAR generation relies on designing viral vectors that can enter the patient’s blood and lymphatic systems. These vectors both infect and activate naïve T helper and T cytotoxic cells into displaying the specifically engineered CAR receptor. The activated T cells with the new CARs then release and expand in the bloodstream engaging with the target cells expressing the target antigen.

Conclusions

Despite the current challenges associated with CAR T cell therapy, such as its side effects, limited practicality and cost, CAR T cell therapy undeniably represents a promising way of affecting immunomodulation. It has the potential to achieve better control and management not only for refractory SLE cases but also for patients with other autoimmune disorders.

At first glance, CAR T cell therapy in autoimmune disorders appears to follow in the footsteps of rituximab, a haemato-oncological treatment that has crossed over to treat autoimmune disorders. This would be an oversight, as the delivery of CAR T cell therapy is far more complex, the side effect profile is more substantial, and the logistical and financial costs are prohibitive. Unlike with the adoption of rituximab, these factors are likely to be a firmer barrier to widespread adoption. Yet, patients with SLE receiving CAR T cell therapy against CD19 have shown clinical and biochemical improvement, a manageable side effect profile and, most importantly, a drug-free sustainable response. This begs the question of whether this deep reset of the immune system via a more extensive B cell depletion can change the fundamental biology of SLE.

Undoubtedly, the challenges related to CAR T cell therapy will be easier to overcome as research on the current generations of CAR T cell therapy continues to improve and evidence to guide strategies for minimizing side effects and improving safety increases.

We are only beginning to scratch the surface of this technology. CAR T cell therapy is finding uses in an increasing list of conditions, starting with haematological malignancies and moving on to solid tumours and, more recently, to autoimmune disorders. It is rare that a new treatment comes along with so much promise and potential. Further research is needed to reveal whether this exciting new therapy will significantly impact the way we manage SLE.

Article Information:
Disclosure

Christopher J Edwards has received advisory board, speaker’s bureau and research support fees from Abbvie, BMS, Biogen, Celgene, Eli Lilly, Fresenius, Galapagos, Gilead, GSK, Janssen, Novartis, Pfizer, Roche, and Sanofi. Michael Alsharkawy, Alice Mason and May Lwin have no financial or non-financial relationships or activities to declare in relation to this article.

Compliance With Ethics

This article involves a review of the literature and did not involve any studies with human or animal subjects performed by any of the authors.

Review Process

Double-blind peer review.

Authorship

The named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship of this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.

Correspondence

Alice MasonRheumatology Department, University Hospital SouthamptonSouthampton, SO16 6YDUKAlice.Mason@uhs.nhs.uk

Support

No funding was received in the publication of this article.

Access

This article is freely accessible at touchIMMUNOLOGY.com © Touch Medical Media 2023

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analysed during the writing of this article.

Received

2023-05-08

References

1. Tsokos GCSystemic lupus erythematosusN Engl J Med2011;365:211021DOI10.1056/NEJMra1100359.

2. Katarzyna PBWiktor SEwa DPiotr LCurrent treatment of systemic lupus erythematosus: A clinician’s perspectiveRheumatol Int2023;43:1395407DOI10.1007/s00296-023-05306-5.

3. Murphy GIsenberg DANew therapies for systemic lupus erythematosus: Past imperfect, future tenseNat Rev Rheumatol2019;15:40312DOI10.1038/s41584-019-0235-5.

4. Fanouriakis ATziolos NBertsias GBoumpas DTUpdate on the diagnosis and management of systemic lupus erythematosusAnn Rheum Dis2021;80:1425DOI10.1136/annrheumdis-2020-218272.

5. Cooper GSDooley MATreadwell ELet alRisk factors for development of systemic lupus erythematosus: Allergies, infections, and family historyJ Clin Epidemiol2002;55:9829DOI10.1016/s0895-4356(02)00429-8.

6. Marion TNPostlethwaite AEChance, genetics, and the heterogeneity of disease and pathogenesis in systemic lupus erythematosusSemin Immunopathol2014;36:495517DOI10.1007/s00281-014-0440-x.

7. Rioux JDGoyette PVyse TJet alMapping of multiple susceptibility variants within the MHC region for 7 immune-mediated diseasesProc Natl Acad Sci U S A2009;106:186805DOI10.1073/pnas.0909307106.

8. Sullivan KEGenetics of systemic lupus erythematosus: Clinical implicationsRheum Dis Clin North Am. 2000;26:22956DOI10.1016/s0889-857x(05)70137-x.

9. Ceccarelli FPerricone CBorgiani Pet alGenetic factors in systemic lupus erythematosus: Contribution to disease phenotypeJ Immunol Res2015;2015:745647DOI10.1155/2015/745647.

10. Iwamoto TNiewold TBGenetics of human lupus nephritisClin Immunol2017;185:329. DOI10.1016/j.clim.2016.09.012.

11. Kotzin BLSystemic lupus erythematosusCell1996;85:3036DOI10.1016/s0092-8674(00)81108-3.

12. Theofilopoulos ANThe basis of autoimmunity: Part I. Mechanisms of aberrant self-recognitionImmunol Today1995;16:908DOI10.1016/0167-5699(95)80095-6.

13. Ohl KTenbrock KRegulatory T cells in systemic lupus erythematosusEur J Immunol2015;45:34455DOI10.1002/eji.201344280.

14. Hahn BHEbling FSingh RRet alCellular and molecular mechanisms of regulation of autoantibody production in lupusAnn N Y Acad Sci2005;1051:43341DOI10.1196/annals.1361.085.

15. Katsuyama TTsokos GCMoulton VRAberrant T cell signaling and subsets in systemic lupus erythematosusFront Immunol2018;9:1088DOI10.3389/fimmu.2018.01088.

16. Cohen-Solal JFGJeganathan VHill Let alHormonal regulation of B-cell function and systemic lupus erythematosusLupus2008;17:52832DOI10.1177/0961203308089402.

17. Lehmann PHölzle EKind Pet alExperimental reproduction of skin lesions in lupus erythematosus by UVA and UVB radiationJ Am Acad Dermatol1990;22:1817DOI10.1016/0190-9622(90)70020-i.

18. Li JMay WMcMurray RWPituitary hormones and systemic lupus erythematosusArthritis Rheum2005;52:370112DOI10.1002/art.21436.

19. Daniel J.WBevra HDubois’ Lupus Erythematosus and Related Syndromes9th edAmsterdam: Elsevier2018.

20. James JARobertson JMLupus and Epstein-BarrCurr Opin Rheumatol2012;24:3838DOI10.1097/BOR.0b013e3283535801.

21. Lahita RGSex and gender influence on immunity and autoimmunityFront Immunol2023;14:1142723. DOI10.3389/fimmu.2023.1142723.

22. McMurray RWMay WSex hormones and systemic lupus erythematosus: Review and meta-analysisArthritis Rheum2003;48:210010DOI10.1002/art.11105.

23. Houen GTrier NHEpstein-barr virus and systemic autoimmune diseasesFront Immunol2020;11:587380. DOI10.3389/fimmu.2020.587380.

24. Grillo-López AJWhite CADallaire BKet alRituximab: The first monoclonal antibody approved for the treatment of lymphomaCurr Pharm Biotechnol2000;1:19DOI10.2174/1389201003379059.

25. Ferraccioli GFDe Vita SCasatta Let alAutoimmune connective tissue disease, chronic polyarthritides and B cell expansion: Risks and perspectives with immunosuppressive drugsClin Exp Rheumatol1996;14 Suppl 14:S7180.

26. Gorman CLeandro MIsenberg DB cell depletion in autoimmune diseaseArthritis Res Ther2003;5 Suppl 4:S1721DOI10.1186/ar1007.

27. Karrar SCunninghame Graham DSAbnormal B cell development in systemic lupus erythematosus: What the genetics tell usArthritis Rheumatol2018;70:496507DOI10.1002/art.40396.

28. Leandro MJEdwards JCWCambridge GClinical outcome in 22 patients with rheumatoid arthritis treated with B lymphocyte depletionAnn Rheum Dis2002;61:8838DOI10.1136/ard.61.10.883.

29. Leandro MJEdwards JCCambridge Get alAn open study of B lymphocyte depletion in systemic lupus erythematosusArthritis Rheum2002;46:26737DOI10.1002/art.10541.

30. Bertsias GIoannidis JPABoletis Jet alEULAR recommendations for the management of systemic lupus erythematosus. Report of a Task Force of the EULAR Standing Committee for International Clinical Studies Including TherapeuticsAnn Rheum Dis2008;67:195205DOI10.1136/ard.2007.070367.

31. Sfikakis PPBoletis JNLionaki Set alRemission of proliferative lupus nephritis following B cell depletion therapy is preceded by down-regulation of the T cell costimulatory molecule CD40 ligand: An open-label trialArthritis Rheum2005;52:50113DOI10.1002/art.20858.

32. Leandro MJCambridge GEdwards JCet alB-cell depletion in the treatment of patients with systemic lupus erythematosus: A longitudinal analysis of 24 patientsRheumatology (Oxford)2005;44:15425DOI10.1093/rheumatology/kei080.

33. Hahn BHMcMahon MAWilkinson Aet alAmerican College of Rheumatology guidelines for screening, treatment, and management of lupus nephritisArthritis Care Res2012;64:797808DOI10.1002/acr.21664.

34. Kostopoulou MFanouriakis ACheema Ket alManagement of lupus nephritis: a systematic literature review informing the 2019 update of the joint EULAR and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendationsRMD Open2020;6:e001263DOI10.1136/rmdopen-2020-001263.

35. Gordon CAmissah-Arthur MBGayed Met alThe British Society for Rheumatology guideline for the management of systemic lupus erythematosus in adultsRheumatology (Oxford)2018;57:e145DOI10.1093/rheumatology/kex286.

36. Hahn BHBelimumab for systemic lupus erythematosusN Engl J Med2013;368:152835DOI: 10.1056/NEJMct1207259.

37. Merrill JJune JKoumpouras Fet alFRI0176 phase II, double-blind, randomized, placebo-controlled study of a reversible B cell inhibitor, Xmab®5871, in systemic lupus erythematosus (SLE)Ann Rheum Dis2019;78:76162DOI10.1136/annrheumdis-2019-eular.4234.

38. Clowse MEBWallace DJFurie RAet alEfficacy and safety of epratuzumab in moderately to severely active systemic lupus erythematosus: Results from two phase III randomized, double-blind, placebo-controlled trialsArthritis Rheumatol2017;69:36275DOI10.1002/art.39856.

39. Isenberg DGordon CLicu Det alEfficacy and safety of atacicept for prevention of flares in patients with moderate-to-severe systemic lupus erythematosus (SLE): 52- week data (APRIL-SLE randomised trial)Ann Rheum Dis2015;74:200615DOI10.1136/annrheumdis-2013-205067.

40. Isenberg DAFurie RAJones Net alEfficacy, safety, and pharmacodynamics effects of the Bruton’s tyrosine kinase inhibitor, fenebrutinib (GDC-0853), in moderate to severe systemic lupus erythematosus: Results of a phase II randomized controlled trialArthritis Rheumatol2019;71.

41. Ishii TTanaka YKawakami Aet alMulticenter double-blind randomized controlled trial to evaluate the effectiveness and safety of bortezomib as a treatment for refractory systemic lupus erythematosusMod Rheumatol2018;28:98692DOI10.1080/14397595.2018.1432331.

42. Bag-Ozbek AHui-Yuen JSEmerging B-cell therapies in systemic lupus erythematosusTher Clin Risk Manag2021;17:3954DOI10.2147/TCRM.S252592.

43. AstraZenecaSaphnelo approved in the EU for the treatment of moderate to severe systemic lupus erythematosus2022Available atwww.astrazeneca.com/media-centre/press-releases/2022/saphnelo-approved-in-eu-for-sle.html#:~:text=AstraZeneca’s%20Saphnelo%20(anifrolumab)%20has%20been,)%2C%20despite%20receiving%20standard%20therapy (Date last accessed29 July 2023).

44. Loncharich MFAnderson CWInterferon inhibition for lupus with anifrolumab: Critical appraisal of the evidence leading to FDA approvalACR Open Rheumatol2022;4:48691DOI10.1002/acr2.11414.

45. Merrill JTNeuwelt CMWallace DJet alEfficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: The randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trialArthritis Rheum2010;62:22233DOI10.1002/art.27233.

46. Barber MRWDrenkard CFalasinnu Tet alGlobal epidemiology of systemic lupus erythematosusNat Rev Rheumatol2021;17:51532DOI10.1038/s41584-021-00690-3.

47. Aringer MAlarcón-Riquelme MEClowse Met alA glimpse into the future of systemic lupus erythematosusTher Adv Musculoskelet Dis2022;14:1759720X221086719DOI10.1177/1759720X221086719.

48. Vormittag PGunn RGhorashian SVeraitch FSA guide to manufacturing CAR T cell therapiesCurr Opin Biotechnol2018;53:16481DOI10.1016/j.copbio.2018.01.025.

49. Schuster SJSvoboda JChong EAet alChimeric antigen receptor T cells in refractory B-cell LymphomasN Engl J Med2017;377:254554DOI10.1056/NEJMoa1708566.

50. Marofi FMotavalli RSafonov VAet alCAR T cells in solid tumors: Challenges and opportunitiesStem Cell Res Ther2021;12:81DOI10.1186/s13287-020-02128-1.

51. Finck AVBlanchard TRoselle CPet alEngineered cellular immunotherapies in cancer and beyondNat Med2022;28:67889DOI10.1038/s41591-022-01765-8.

52. Gross GWaks TEshhar ZExpression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificityProc Natl Acad Sci U S A1989;86:100248DOI10.1073/pnas.86.24.10024.

53. Levine BLMiskin JWonnacott KKeir CGlobal manufacturing of CAR T cell therapyMol Ther Methods Clin Dev2017;4:92101DOI10.1016/j.omtm.2016.12.006.

54. Fesnak AO’Doherty UClinical development and manufacture of chimeric antigen receptor T cells and the role of leukapheresisEur Oncol Haematol2017;13:28DOI10.17925/EOH.2017.13.01.28.

55.U.S. Food and Drug Administration.Package insert KYMRIAH2023Available at: www.fda.gov/media/107296/download (Date last accessed1 May 2023).

56.U.S. Food and Drug Administration.Package insert CARVYKTI®2023Available at: www.fda.gov/media/156560/download (Date last accessed1 May 2023).

57. U.S. Food and Drug AdministrationVaccines, blood & biologicsAvailable at: www.fda.gov/vaccines-blood-biologics (Date last accessed29 March 2023).

58. European medicines agencyNews and events2023Available at: www.ema.europa.eu/en (Date last accessed25 April 2023).

59.NHS England.CAR-T therapyAvailable atwww.england.nhs.uk/cancer/cdf/car-t-therapy/ (Date last accessed29 March 2023).

60. National Institute for Health and Care ExcellenceTwo new personalised immunotherapy treatments for aggressive forms of blood cancer recommended for the cancer drugs fund2023Available atwww.nice.org.uk/news/article/two-new-personalised-immunotherapy-treatments-for-aggressive-forms-of-blood-cancer-recommended-for-the-cancer-drugs-fund (Date last accessed31 July 2023).

61. Kansal RRichardson NNeeli Iet alSustained B cell depletion by CD19-targeted CAR T cells is a highly effective treatment for murine lupusSci Transl Med2019;11:eaav1648DOI10.1126/scitranslmed.aav1648.

62. Jin XXu QPu Cet alTherapeutic efficacy of anti-CD19 CAR-T cells in a mouse model of systemic lupus erythematosusCell Mol Immunol2021;18:1896903DOI10.1038/s41423-020-0472-1.

63. Zhang WFeng JCinquina Aet alTreatment of systemic lupus erythematosus using BCMA-CD19 compoundStem Cell Rev and Rep2021;17:21203DOI10.1007/s12015-021-10251-6.

64. Mougiakakos DKrönke GVölkl Set alCD19-targeted CAR T cells in refractory systemic lupus erythematosusN Engl J Med2021;385:5679DOI10.1056/NEJMc2107725.

65. Mackensen AMüller FMougiakakos Det alAnti-CD19 CAR T cell therapy for refractory systemic lupus erythematosusNat Med2022;28:212432DOI10.1038/s41591-022-02017-5.

66. Reddy VCambridge GIsenberg DAet alInternalization of rituximab and the efficiency of B cell depletion in rheumatoid arthritis and systemic lupus erythematosusArthritis Rheumatol2015;67:204655DOI10.1002/art.39167.

67. Kamburova EGKoenen HJPMBorgman KJEet alA single dose of rituximab does not deplete B cells in secondary lymphoid organs but alters phenotype and functionAm J Transplant2013;13:150311DOI10.1111/ajt.12220.

68. Aringer MAlarcón-Riquelme MEClowse Met alA glimpse into the future of systemic lupus erythematosusTher Adv Musculoskelet Dis2022;14:1759720X221086719DOI10.1177/1759720X221086719.

69. Curran EO’Brien MRole of blinatumomab, inotuzumab, and CAR T-cells: Which to choose and how to sequence for patients with relapsed diseaseSemin Hematol2020;57:15763DOI10.1053/j.seminhematol.2020.11.001.

70. Sermer DBatlevi CPalomba MLet alOutcomes in patients with DLBCL treated with commercial CAR T cells compared with alternate therapiesBlood Adv2020;4:466978DOI10.1182/bloodadvances.2020002118.

71. Regazzi MBIacona IAvanzini MAet alPharmacokinetic behavior of rituximab: A study of different schedules of administration for heterogeneous clinical settingsTher Drug Monit2005;27:78592DOI10.1097/01.ftd.0000184162.60197.c1.

72. Oomen INassar-Sheikh Rashid ABouts AHMet alAnti-rituximab antibodies affect pharmacokinetics and pharmacodynamics of rituximab in children with immune-mediated diseasesClin Exp Rheumatol2022;40:18390DOI10.55563/clinexprheumatol/ftira8.

73. McLellan ADAli Hosseini Rad SMChimeric antigen receptor T cell persistence and memory cell formationImmunol Cell Biol2019;97:66474DOI10.1111/imcb.12254.

74. Albinger NHartmann JUllrich ECurrent status and perspective of CAR-T and CAR-NK cell therapy trials in GermanyGene Ther2021;28:51327DOI10.1038/s41434-021-00246-w.

75. Grupp SAKalos MBarrett Det alChimeric antigen receptor-modified T cells for acute lymphoid leukemiaN Engl J Med2013;368:150918DOI10.1056/NEJMoa1215134.

76. Neelapu SSLocke FLBartlett NLet alAxicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphomaN Engl J Med2017;377:253144DOI10.1056/NEJMoa1707447.

77. Lin HCheng JMu Wet alAdvances in universal CAR-T cell therapyFront Immunol2021;12:744823DOI: 10.3389/fimmu.2021.744823.

78. Sterner RCSterner RMCAR-T cell therapy: Current limitations and potential strategiesBlood Cancer J2021;11:69DOI10.1038/s41408-021-00459-7.

79. Markouli MUllah FUnlu Set alToxicity profile of chimeric antigen receptor T-cell and bispecific antibody therapies in multiple myeloma: Pathogenesis, prevention and managementCurr Oncol2023;30:633052DOI10.3390/curroncol30070467.

80. GenentechFDA approves Genentech’s Actemra (tocilizumab) for the treatment of CAR T cell-induced cytokine release syndrome2017Available atwww.gene.com/media/press-releases/14679/2017-08-30/fda-approves-genentechs-actemra-tocilizu (Date last accessed30 July 2023).

81. Schubert M-LSchmitt MWang Let alSide-effect management of chimeric antigen receptor (CAR) T-cell therapyAnn Oncol2021;32:3448DOI10.1016/j.annonc.2020.10.478.

82. Lee DWSantomasso BDLocke FLet alASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cellsBiol Blood Marrow Transplant2019;25:62538DOI10.1016/j.bbmt.2018.12.758.

83. Pasqui DMLatorraca CPacheco RLRiera RCAR-T cell therapy for patients with hematological malignancies. A systematic reviewEur J Haematol2022;109:60118DOI10.1111/ejh.13851.

84. Ernst MOeser ABesiroglu Bet alChimeric antigen receptor (CAR) T-cell therapy for people with relapsed or refractory diffuse large B-cell lymphomaCochrane Database Syst Rev2021;9:CD013365DOI10.1002/14651858.CD013365.pub2.

85. Kampouri EWalti CSGauthier JHill JAManaging hypogammaglobulinemia in patients treated with CAR-T-cell therapy: Key points for cliniciansExpert Rev Hematol2022;15:30520DOI10.1080/17474086.2022.2063833.

86. Wiedmeier-Nutor JEIqbal MRosenthal ACet alResponse to COVID-19 vaccination post-CAR T therapy in patients with non-Hodgkin lymphoma and multiple myelomaClin Lymphoma Myeloma Leuk2023;23:45662DOI10.1016/j.clml.2023.03.002.

87. Aleissa MMLittle JSDavey Set alSARS-cov-2 vaccine immunogenicity among chimeric antigen receptor T-cell therapy recipientsTransplant Cell Ther2023;6367:011636.

88. Goldman ARaschi EChapman Jet alProgressive multifocal leukoencephalopathy in patients treated with chimeric antigen receptor T cellsBlood2023;141:6737DOI10.1182/blood.2022017386.

89. Berger JRMalik VLacey Set alProgressive multifocal leukoencephalopathy in rituximab-treated rheumatic diseases: A rare eventJ Neurovirol2018;24:32331DOI10.1007/s13365-018-0615-7.

90. Cellular Therapy Summary Slides 2021Center for International blood and marrow transplant research2022Available athttps://cibmtr.org/CIBMTR/Resources/Summary-Slides-Reports (Date last accessed30 July 2023).

91. Shah NNAhn KWLitovich Cet alIs autologous transplant in relapsed DLBCL patients achieving only a PET+ PR appropriate in the CAR T-cell era Blood2021;137:141623DOI10.1182/blood.2020007939.

92. Kretschmann SVölkl SReimann Het alSuccessful generation of CD19 chimeric antigen receptor T cells from patients with advanced systemic lupus erythematosusTransplant Cell Ther2023;29:2733DOI10.1016/j.jtct.2022.10.004.

93. National Institute for Health and Care ExcellenceAxicabtagene ciloleucel for treating diffuse large B-cell lymphoma and primary mediastinal large B-cell lymphoma after 2 or more systemic therapiesAvailable atwww.nice.org.uk/guidance/ta872/chapter/2-Information-about-axicabtagene-ciloleucel (Date last accessed30 March 2023).

94. Labanieh LMackall CLCAR immune cells: Design principles, resistance and the next generation. Nature2023;619:63548. DOI: 10.1038/s41586-023-06088-3.

95. Muthuvel MSrinivasan HLouis LMartin SEngineering off-the-shelf universal CAR T cells: A silver lining in the cloudCytokine2022;156:155920DOI10.1016/j.cyto.2022.155920.

96. Young RMEngel NWUslu Uet alNext-generation CAR T-cell therapiesCancer Discov2022;12:162533DOI10.1158/2159-8290.CD-21-1683.

97. Arias JYu JVarshney Met alHematopoietic stem cell- and induced pluripotent stem cell-derived CAR-NK cells as reliable cell-based therapy solutionsStem Cells Transl Med2021;10:98795DOI10.1002/sctm.20-0459.

98. Michels AHo NBuchholz CJPrecision medicine: In vivo CAR therapy as a showcase for receptor-targeted vector platformsMol Ther2022;30:240115DOI10.1016/j.ymthe.2022.05.018.

Further Resources

Share this Article
Related Content In Systemic Lupus Erythematosus
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72