Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Biomedical Technology 4 (2023) 1–10

Contents lists available at ScienceDirect

Biomedical Technology
journal homepage: www.keaipublishing.com/en/journals/biomedical-technology

Review

Developing engineering technologies for the treatment of systemic


lupus erythematosus
Xubin Hao a, Yuze Wang a, Rui Liu a, Dagan Zhang a, *, Bin Kong a, b, **, Jun Liang a, ***,
Lingyun Sun a
a
Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing,
210008, China
b
Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China

A R T I C L E I N F O A B S T R A C T

Keywords: Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects multiple organs and tissues.
Engineering technology However, only a handful of new drugs have been FDA-approved for SLE since the 1950s. Therefore, novel
Systemic lupus erythematous treatments for SLE are urgently needed to be developed. In recent years, various engineering technologies such as
Nanocarrier
tissue engineering, organs-on-chips, and intelligent delivery systems have been rapidly developed in the field of
Drug delivery
biomedicine. Notably, engineered nanocarriers and cell-based therapies can address the problems faced by
Treatment
traditional drug delivery and cell transplantation. They have proven effective in the treatment of many areas of
disease, including autoimmune diseases. This is an important opportunity to break through the limited treatment
options for SLE. In this review, we summarize the application progresses of engineering technologies and also
propose their challenges in SLE treatment. This paper aims to help readers to understand the perspective of
engineering technologies on the direction of SLE treatments in forthcoming years.

1. Introduction [9,10]. Hence, the development of new treatments for SLE cannot wait
any longer.
Systemic lupus erythematosus (SLE), also known as lupus, is a het- In recent years, the development of engineering technology has given
erogeneous autoimmune disease characterized by chronic inflammation birth to biomedical engineering. Tissue engineering, intelligent delivery
[1]. Patients with lupus lose immune tolerance and produce numerous systems, organs-on-chips, etc. shine in many fields [11–13]. Among
autoantibodies that attack their healthy tissues [2]. If not treated in time, them, engineered nanocarriers and cell-based therapies have been
the disease will recur and cause irreversible damage to the affected or- rapidly applied and developed in various diseases, including autoim-
gans, eventually leading to the death of the patient [3]. Although much of mune diseases [14,15]. Nanocarriers commonly used for disease treat-
the pathophysiology of SLE has been uncovered in recent years, sup- ment include synthetic liposomes, polymeric nanoparticles, biologically
pression of disease activity and reduction of organ damage remain the derived cell membrane (CM)-coated nanoparticles, and extracellular
mainstay of lupus management by rheumatologists [4]. Medications vesicles (EVs). Nanocarriers can improve the solubility and accumulation
commonly used to treat SLE include glucocorticoids, nonsteroidal of traditional drugs in inflamed tissues through enhanced permeability
anti-inflammatory drugs (NSAIDs), antimalarials, and immunosuppres- and retention (EPR) effects [16], or enable active targeting through
sants [5,6]. However, these drugs have poor specificity, and long-term surface modification [17]. When combined with CM coating technology,
use will be accompanied by severe or even life-threatening adverse re- nanocarriers can achieve long blood circulation and inherit some specific
actions [7]. Encouragingly, the advent of belimumab in 2011 broke the functions of the source cells [18]. Besides, the development of engi-
plight of no new drug available for lupus patients for nearly 60 years [8]. neering technology has also led to engineered cell growth. In the treat-
However, belimumab has narrow indications and some adverse reactions ment of SLE, chimeric antigen receptor-T (CAR-T) cells and engineered

* Corresponding author.
** Corresponding author. Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing
University Medical School, Nanjing, 210008, China.
*** Corresponding author.
E-mail addresses: zhang_dagan@126.com (D. Zhang), kongbin13@163.com (B. Kong), 13505193169@139.com (J. Liang).

https://doi.org/10.1016/j.bmt.2023.02.002
Received 30 January 2023; Received in revised form 16 February 2023; Accepted 19 February 2023
Available online 22 March 2023
2949-723X/© 2023 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
X. Hao et al. Biomedical Technology 4 (2023) 1–10

mesenchymal stem cells (MSCs) have been explored [19,20]. Preclinical and immunosuppressive properties of glucocorticoids make them useful
and clinical achievements of CAR-T cell therapy in refractory SLE are in both quiescent and active phases of disease [23]. However, gluco-
encouraging. Engineered MSCs can enhance the stability of MSCs corticoids have many notorious adverse events, including affecting
transplantation therapy and provide other routes for MSCs trans- metabolism, bone density, and increasing the risk of infection, especially
plantation to solve the problem of intravenous infusion [21]. for long-term use [7]. Despite widespread acceptance that glucocorti-
In this review, we first introduce the main drugs for the clinical coids have a beneficial effect on suppressing inflammation in SLE, several
treatment of SLE and their shortcomings. Subsequently, we elaborate on studies have also shown that long-term glucocorticoid therapy for
the development and achievements of engineering technologies applied maintaining remission is connected with a higher chance of organ
to SLE therapies. The engineering technologies mainly include nano- damage [24]. Therefore, whether to discontinue glucocorticoids in pa-
carriers and cell-based therapies (Fig. 1). Finally, the prospects of engi- tients with lupus in remission still needs to be explored [25].
neering technologies in the field of SLE treatment are discussed.
Therefore, this review is expected to provide comprehensive and helpful
2.2. NSAIDs
information for researchers exploring better therapies for SLE.

NSAIDs are commonly used to manage joint pain, fever, serositis, and
2. Current clinical therapeutic approaches
headache in patients with SLE [26]. NSAIDs inhibit the cyclooxygenase
(COX) enzymes (COX-1 and COX-2), which are involved in the produc-
2.1. Glucocorticoids
tion of prostaglandins [27]. While non-selective NSAIDs exert analgesic
effects, they can also cause adverse gastrointestinal and kidney reactions.
Glucocorticoids are still first-line options and have been used to treat
Selective COX-2 inhibitors such as celecoxib are safer for the gastroin-
SLE for more than 60 years [22]. The potent and rapid anti-inflammatory
testinal tract but can increase the risk to the cardiovascular system.

Fig. 1. Schematic illustration of engineering technologies for SLE treatment.

2
X. Hao et al. Biomedical Technology 4 (2023) 1–10

Therefore, the type and dose of NSAIDs must be individualized according [43]. Thus, they fabricated polymeric nanoparticles conjugated to a small
to the patient's gastrointestinal and cardiovascular risk stratification molecule gambogic acid (P2Ns-GA) to increase the solubility and tar-
[28]. geting of CsA. Gambogic acid is a ligand targeting CD71, which is
expressed by the intestinal epithelia, Peyer's patches, and lymphocytes.
2.3. Antimalarials After incubation with peripheral blood mononuclear cells (PBMCs) in
vitro for 1 h, P2Ns-GA were internalized whereas P2Ns were mostly
Antimalarial drugs are the cornerstone of lupus treatment [29]. The constricted to the cell surface (Fig. 2a). In vivo drug distribution experi-
first use of antimalarials in SLE patients probably dates back to 1894, ments showed that the concentration of P2Ns-GA-CsA in lymph nodes
when J. S. Payne described the successful treatment of a lupus rash with was 18 times higher than that of free CsA group. Eventually, after
quinine. During World War II, it was found that quinacrine, used to P2Ns-GA-CsA treatment, various disease indicators of lupus mice were
prevent malaria, could relieve some of the symptoms of soldiers suffering significantly improved [44]. Jiang et al. prepared mycophenolic acid
from rheumatism, which led to research on antimalarial drugs for (MPA)-loaded dextran (MPA@Dex-MPA) nanoparticles to improve the
rheumatic disease [30]. Later, in the 1950s, chloroquine and hydroxy- pharmacokinetics of MPA. It was found that MPA@Dex-MPA nano-
chloroquine came out one after another and have been used for treating particles mainly accumulated in the spleen and kidney and were
SLE for more than 60 years. Nowadays, hydroxychloroquine is accepted phagocytized by macrophages. After treatment, macrophages were
globally for SLE patients without contraindications, even in lupus significantly reduced, predominantly exhibiting an M2-like phenotype. T
nephritis patients and pregnant women [31,32]. Of course, retinopathy, cell proliferation was also inhibited in the spleen and kidney (Fig. 2b). All
gastrointestinal discomfort, and cutaneous reactions still need attention the results indicated that the MPA@Dex-MPA nanoparticles relieved the
[33]. disease progression in MRL/lpr mice [45]. Azathioprine is commonly
used to treat severe SLE but has low solubility and a short half-life. Hu
2.4. Immunosuppressants et al. developed polyhydroxyalkanoate (PHA) nanoparticles encapsu-
lating azathioprine (AZA-PHA) for SLE therapy. AZA-PHA nanoparticles
When lupus is severe or vital organs are involved, immunosuppres- exhibited slower clearance and more accumulation in the spleen. As a
sive agents are often utilized with glucocorticoids to control the disease result, AZA-PHA nanoparticles reduced urinary protein with less toxicity
activity. For III and IV lupus nephritis, cyclophosphamide, mycopheno- [46]. Besides immunosuppressants, nanocarriers have also been devel-
late mofetil (MMF), and azathioprine are often used as first-line thera- oped to deliver RNAs or some cytokines for the treatment of lupus [47].
peutic drugs [34]. A similar medication tactic is also used in patients with MicroRNAs (miRNAs) are endogenous noncoding RNAs that regulate
neuropsychiatric SLE (NPSLE) [35]. New immunosuppressants such as mRNA stability and translation. Collecting reports disclosed that miRNA
leflunomide, cyclosporine, and tacrolimus have also been gradually dysfunction is associated with the immune disorder in SLE. For example,
introduced into the treatment of SLE, providing more alternatives for miRNA-125a is able to stabilize regulatory T cell-mediated self-tolerance.
treatment [36]. Encouragingly, in January 2021, voclosporin, an oral However, it was found to be down-regulated in peripheral T cells of
calcineurin inhibitor immunosuppressant, was approved by the FDA in people with SLE [48]. Zhang et al. constructed polymeric nanoparticles to
combination with MMF for adults with active lupus nephritis [37,38]. protect miRNA-125a from degradation and deliver it to splenic T cells
However, immunosuppressants have a wide range of effects and poor (Fig. 2c). The results showed that miRNA-125a encapsulated nano-
targeting, leading to side effects such as infection and organ damage. particles could alleviate lupus by improving the disorder between
effector and regulatory T cells [49].
2.5. Biologic agents
3.1.2. Micelles
In recent years, with a better understanding of the pathogenesis of Micelles are small sub-50nm entities self-assembled from water-
SLE, many biologics have been developed to treat lupus. In 2011, the FDA soluble amphiphilic surfactants and are frequently served for drug de-
approved belimumab for the treatment of lupus, which is the first new livery [50]. Long-term use of glucocorticoids in lupus patients may lead
drug since hydroxychloroquine's approval for lupus in 1955 [8]. In July to osteoporosis and infection. Jia et al. fabricated a poly(ethylene glycol)
2021, the FDA approved anifrolumab, a human monoclonal antibody to (PEG)-based dexamethasone prodrug (ZSJ-0228) to address this chal-
type I interferon receptor subunit 1, for treating adult patients with lenge. The pronounced amphiphilicity of the ZSJ-0228 enables it to
moderate-to-severe SLE [39]. Many other biologics in development for spontaneously self-assemble into micelles in aqueous media, thereby
the treatment of lupus hold great potential. Regarding engineered bio- increasing the water solubility of dexamethasone. As nanocarriers have a
logical agents, many excellent articles have been summarized before [6, natural tendency to accumulate in inflamed tissues through vascular
40], so they are not discussed in this review. leakage [51], the prodrug micelles were found to accumulate in the
inflamed kidney (Fig. 2d). Unlike the results of daily dexamethasone
3. Engineering technologies-based treatment of SLE treatment, monthly ZSJ-0228 treatment greatly relieved lupus nephritis
without significant glucocorticoid-related side effects [52]. Next, the
3.1. Nanocarriers authors went on to investigate the optimal dose-response relationship of
ZSJ-0228 in lupus nephritis. ZSJ-0228 was injected monthly at a dosage
Nanocarriers commonly used in lupus treatment research mainly ranging from 0.5 to 8.0 mg/kg/day dexamethasone equivalent. Urine
include polymeric nanoparticles, micelles, liposomes, and nanogels. protein and other results showed that the most effective dose range for
ZSJ-0228 in NZB/W F1 mice is between 1.0 and 3.0 mg/kg/day dexa-
3.1.1. Polymeric nanoparticles methasone equivalent (Fig. 2e) [53]. Due to fewer side effects and less
Polymeric nanoparticles are widely used for drug delivery due to their frequent dosing, ZSJ-0228 may become an alternative to glucocorticoids
excellent biocompatibility, degradability, and sustained release proper- in patients with SLE.
ties [41]. Polymeric nanoparticles are the most studied nanomaterials for
lupus therapy compared to other nanocarriers. Immunosuppressants can 3.1.3. Liposomes
be targeted for delivery through polymeric nanoparticles to address their Liposomes are nanovesicles self-assembled from phospholipids. The
high non-response rates and adverse effects [42]. Recently, Ganugula amphipathic properties of liposomes make them ideal carriers for both
et al. developed oral cyclosporine A (CsA)-loaded nanoparticles for the hydrophilic and lipophilic drugs, as well as some therapeutic nucleic
lymphatic system targeting to treat lupus. Due to high lipid solubility, acids [54]. The US FDA first approved liposomal doxorubicin in 1995 for
low permeability, and nephrotoxicity, the use of CsA in lupus is limited the clinical treatment of cancer [55]. Like polymeric nanoparticles,

3
X. Hao et al. Biomedical Technology 4 (2023) 1–10

Fig. 2. (a) P2Ns-GA targeted to the lymphatic system. (i). Chemical structure of the P2s-GA polymer. (ii) Confocal imaging of mouse PBMCs incubated with fluo-
rescent (green) P2Ns-GA or P2Ns for 60 min. CD71 (purple), CD3þ T cells (red), and CD20þ B cells (red). Reproduced with permission from Ref. [44] Copyright 2020
The Authors. (b) MPA@Dex-MPA nanoparticles targeted to the spleen and kidney. The scheme of the (i) fabrication and (ii) application of MPA@Dex-MPA nano-
particles. Reproduced with permission from Ref. [45] Copyright 2022 The Authors. (c) Schematic illustration of the microRNA-125a-loaded polymeric nanoparticles
restoring effector/regulatory T Cells balance. Reproduced with permission from Ref. [49] Copyright 2020 American Chemical Society. (d) Micelle-forming dexa-
methasone prodrug reduced glucocorticoid side effects. (i) Structure of PEG-based amphiphilic dexamethasone prodrug ZSJ-0228. (ii) ZSJ-0228 was mainly
distributed in the inflamed kidney Reproduced with permission from Ref. [52] Copyright 2018 American Chemical Society. (e) A dose-escalation design to establish
the optimal dose-response relationships for ZSJ-0228 in treating lupus nephritis. Reproduced with permission from Ref. [53] Copyright 2021 American Chemi-
cal Society.

liposomes have many applications in lupus treatment research. As receptors (LXRs) agonist regulating both the phagocytic activity and in-
glomerular mesangial cells are vital cells involved in lupus nephritis, flammatory response in macrophages. The authors found that
Scindia et al. developed novel immunoliposomes for renal glomerulus PS-lipos-AuNC@T0901317 enhanced the removal of apoptotic cells,
targeting. They noticed that α8 integrin was expressed in the glomeruli of leading to reduced production of anti-dsDNA autoantibodies and in-
normal and nephritic mice. Thus, they built liposomes conjugated with flammatory response [61]. These results provide another idea to improve
anti-α8 integrin for mesangial-targeted delivery. Anti-α8 integrin lipo- the clearance of self-antigens by apoptotic cells to treat lupus, which is
somes discovered in glomeruli and mesangial cells revealed that this different from the delivery of glucocorticoids or immunosuppressants by
novel liposomal delivery system provides a targeted strategy for lupus most nanocarriers. Exaggerated cooperation of autoreactive B cells and
nephritis therapy [56]. Moallem et al. constructed a liposomal steroid CD4þ T helper (Th) leading to autoantibody production is a challenge for
formulation for inflamed tissues targeting through EPR effects to treat lupus treatment [62]. Targeting the SLE-specific upregulation of cos-
MRL/lpr mice [57]. Trans-activating transcriptional activator (TAT) timulatory molecules ICOS or CD40L on Th can block Th-B activation,
peptides are frequently utilized for facilitating cellular uptake [58]. Diao but the current blockade of a single costimulatory molecule is ineffective.
et al. fabricated TAT peptide-cationic liposomes to co-delivery dihy- Zhang et al. fabricated rapamycin-encapsulated costimulatory
droartemisinin (DHA) and high-mobility group box 1 (HMGB1) siRNA ICOS/CD40L-bispecific liposomal nanoparticles to specifically target SLE
(TAT-CLs-DHA/siRNA) for SLE treatment (Fig. 3a). Hyperactivation of Th cells (Fig. 3c). The results exhibited that these bispecific nanoparticles
the Toll-like receptor-4 (TLR4) signal pathway is closely associated with effectively inhibited Th–B cell activation and attenuated SLE progression
lupus nephritis [59]. TLR4 is the primary receptor in the inflammatory [63].
cascade of HMGB1. The results revealed that TAT-CLs-DHA/siRNA
significantly reduced the expression of TLR4 and effectively alleviated 3.1.4. Nanogels
lupus nephritis [60]. Phagocytes in lupus patients have inefficient Nanogels are nanosized crosslinked hydrophilic polymeric networks
removal of apoptotic cells, leading to the assemblage of self-antigens, that are ideal carriers for drug and nucleic acid delivery [64]. The
which eventually induce autoantibodies to attack healthy tissues. Xu application of nanogels in the treatment of SLE was mainly studied by
et al. designed a phosphatidylserine (PS) conjugated liposome-coated Fahmy and colleagues, who mainly explored the delivery of the immu-
gold nanocage (AuNC) encapsulating T0901317 (PS-lipo- nosuppressant MPA to dendritic cells (DCs) by nanogels to alleviate lupus
s-AuNC@T0901317) to treat SLE (Fig. 3b). T0901317 is a liver X nephritis (Fig. 3d). First, they found that the MPA-loaded nanogels

4
X. Hao et al. Biomedical Technology 4 (2023) 1–10

Fig. 3. (a) Structure and treatment strategy of TAT-CLs-DHA/siRNA. Reproduced with permission from Ref. [60] Copyright 2019 The Authors. (b) Schematic
illustration of the preparation procedure of PS-lipos-AuNC@T0901317. Reproduced with permission from Ref. [61] Copyright 2019 Elsevier Ltd. (c) Schematic
representation of ICOS/CD40L bispecific RAP nanoparticles for SLE treatment. Reproduced with permission from Ref. [63] Copyright 2022 Elsevier Ltd. (d) Schematic
illustration of the MPA-loaded nanogels. Reproduced with permission from Ref. [65] Copyright 2013 The American Society for Clinical Investigation.

prolonged survival and attenuated lupus nephritis in NZB/W F1 mice kill target cells by recognizing specific antigens. CAR-T cell therapy was
compared with free MPA [65]. They then revealed that the nanogels first approved by the FDA in 2017 for its efficacy against CD19þ tumors
efficiently internalized by DCs and inhibited the maturation of DCs [69]. CD19 is expressed in almost all stages of B cells [70], the most
characterized by the downregulation of costimulatory molecules and important cells in the pathogenesis of SLE. Therefore, a series of break-
reduction of inflammatory factors. DCs are important in regulating im- throughs of CD19-targeted CAR-T cells in lymphoma has attracted re-
munity and tolerance and play an important role in the pathogenesis of searchers to introduce CAR-T into SLE. CAR-T cell therapy for SLE was
lupus [66]. The authors also constructed CD4-targeted MPA-loaded first studied by Kansal and colleagues in two different mouse models of
nanogels to explore whether they could inhibit the proliferation of T lupus in 2019. They constructed CD19-targeted CAR-T cells based on
cells. The results showed a decrease in the number of CD4þ T cells in purified CD8þ T cells and demonstrated effective and long-lasting
lupus mice, but the overall efficacy was similar to that of non-targeted depletion of B cells following CAR-T cell therapy [71]. In parallel, Jin
nanogels [65]. Efficacious internalization is an important factor for the et al. prepared CD19-targeted CAR-T cells based on total T cells to treat
function of nanogels, so the authors next went on to investigate whether lupus in MRL/lpr mice (Fig. 4a). They found that infusion of CAR-T cell
other classes of nanomaterials can also be effective. They compared the therapy can effectively eliminate B cells and prolong survival both before
most commonly used nanocarrier poly (lactic-co-glycolic acid) (PLGA) and after the onset of the disease [70]. The experiments of the above two
nanoparticles. They found that nanogels were more efficiently internal- different teams were all carried out on lupus mice, which provided a basis
ized by dendritic cells and inhibited dendritic cell maturation in vitro. for CAR-T application in lupus patients. Coincidentally, in August 2021,
These results displayed that the ease of internalization of nanogels is an another two teams successively reported the successful application of
important factor for their therapeutic efficacy compared with other CAR-T therapy in the treatment of lupus patients. First, Mougiakakos
nanomaterials [67]. et al. reported that they developed CD19-targeted CAR-T cells to treat a
20-year-old woman with refractory SLE with type IIIA nephritis (Fig. 4b).
3.2. Cell-based therapies The woman did not respond obviously to conventional medical therapy
as well as novel biologics including belimumab and rituximab. After
The application of cell-based therapies or carriers for SLE treatment suspending all treatment except low-dose glucocorticoids, the woman
generally includes four aspects: CAR-T cell therapy, engineered MSCs, was treated with CAR-T cells [72]. Encouragingly, the results showed
CM-coated nanoparticles, and engineered EVs. that the SLE Disease Activity Index (SLEDAI) of this woman returned to
0 [19]. Similarly, Zhang et al. reported that a case of SLE with diffuse
3.2.1. CAR-T large B-cell lymphoma was successfully treated with CD19-BCMA com-
CAR-T cells are T cells engineered to express a CAR consisting of an pound CAR-T cell therapy [73]. Both reports above were based on indi-
extracellular antigen-recognizing domain and an intracellular T cell- vidual case studies but still revealed that CAR-T cell therapy holds huge
activation domain [68]. When T cells are armed with CAR, they can promise in SLE therapy. Recently, CAR-T cell therapy was used in 5

5
X. Hao et al. Biomedical Technology 4 (2023) 1–10

Fig. 4. (a) Working scheme of CAR-T cell manufacture and adoptive transfer in a mouse experiment. Reproduced with permission from Ref. [70] Copyright 2020 CSI
and USTC. (b) Workflow of anti-CD19 CAR-T cell treatment for the SLE patient. Reproduced with permission from Ref. [19] Copyright 2021 The Authors. (c)
Schematic demonstrating the fabrication of adhesive porous particles with MSCs encapsulation for immunomodulation after intraperitoneal transplantation in MRL/lpr
mice. Reproduced with permission from Ref. [21] Copyright 2020 The Authors. (d) Fabrication of the microgels with human MSCs encapsulation via microfluidic
electrospray for immunoregulation after subcutaneous transplantation into MRL/lpr mice. Reproduced with permission from Ref. [20] Copyright 2022 Elsevier Ltd.

young SLE patients with multiorgan involvement. As in other studies, enhanced immunosuppression and reduced systemic inflammation in
glucocorticoids, immunosuppressants, and biologics were ineffective in lupus mice [80]. Another factor affecting the clinical efficacy of MSCs
these 5 patients. Three months after a single infusion of CAR-T cells, the therapy is that after intravenous infusion, MSCs are easily lysed by im-
average SLEDAI of all patients was 0. Furthermore, drug-free remission mune cells and maintain a short time in vivo [81,82]. To overcome this
was maintained during a median follow-up of 8 months [74]. Unlike problem, Zhao and colleagues recently synthesized microparticles and
individual case studies, the results of these 5 patients make the applica- microgels to encapsulate MSCs by microfluidic electrospray technology
tion of CAR-T in lupus promising to be a study with a larger sample. for treating lupus mice [20,21]. First, inspired by the natural micro-
structure of the sand tower worm nest, they fabricated MSCs encapsu-
3.2.2. Engineered MSCs lated adhesive porous particles (Fig. 4c). After intraperitoneal injection,
MSCs are fibroblast-like pluripotent stem cells without hematopoietic compared with unencapsulated MSCs, microsphere encapsulation can
function [75]. They can modulate innate and adaptive immunity for prolong the existence time of MSCs in vivo, which is of great significance
immunomodulatory functions. Therefore, MSC transplantation has been for exerting the therapeutic effect of MSCs. Meanwhile, MSCs encapsu-
introduced to treat SLE. Both preclinical and clinical studies have lated microparticles induced macrophages to shift to the M2 phenotype
revealed that allogeneic MSC transplantation could effectively relieve the and powerfully relieved lupus nephritis [21]. Similarly, subcutaneous
clinical symptoms of patients with refractory lupus [76,77]. However, injection of microgels encapsulated with MSCs also had a good effect on
some studies have shown that MSC transplantation is ineffective, which MRL/lpr mice (Fig. 4d) [20]. These results provide some new delivery
may be related to the immune regulation elasticity of MSCs and the approaches for MSCs transplantation beyond just intravenous infusion.
susceptibility to the microenvironment [78]. Hence, engineered MSCs
such as genetic modification can compensate for and increase the func- 3.2.3. CM-coated nanoparticles
tion of MSCs. Che et al. found that lupus bone marrow MSCs exhibit Although nanocarriers have achieved a series of achievements in pre-
functional deficits in normal B-cell suppression compared with normal clinical and clinical studies, there are still some areas for improvement. To
bone marrow MSCs. They further explored that knocking down CCL2 in avoid immune clearance, the surface of nanocarriers usually needs to be
normal MSCs disrupted their ability to suppress B cells. Thus, they con- modified with PEG to improve blood circulation time. However, PEGyla-
structed engineered lupus MSCs overexpressing CCL2, which effectively tion will induce the body to produce antibodies, which will accelerate the
restored the ability of MSCs to regulate B cells and alleviated lupus body's clearance of nanocarriers after multiple injections [83]. In addition,
nephritis [79]. Likewise, genetically modified MSCs overexpressing IL-37 it is often necessary to incorporate some ligands to increase the targeting

6
X. Hao et al. Biomedical Technology 4 (2023) 1–10

of nanocarriers, but the characterization and preparation of ligands are 3.2.4. Engineered EVs
complicated. To improve these deficiencies, in 2011, Zhang and col- EVs are lipid bilayer vesicles secreted by various cells for cellular
leagues proposed for the first time to camouflage nanocarriers by coating communication in vivo. Unlike simple liposomes, EVs contain complex
PLGA nanoparticles with red blood cell (RBC) membrane [84]. The blood lipids, nucleic acids, and proteins [87]. The most generally studied EVs
circulation experiments showed that the elimination half-life of are exosomes, 40–100 nm in diameter. Exosomes themselves can regu-
RBC-coated nanoparticles was 39.6 h, which was longer than that of late almost all immune cells (Fig. 5c). At the same time, the vesicle
PEGylated nanoparticles (15.8 h). Since then, CM coating technology structure allows them to be used as nanocarriers to deliver nucleic acids
derived from different types of cells has been rapidly applied in cancer and or drugs [88]. Exosomes have been proven to play a critical role in the
other fields. However, the application of cell membrane coating technol- pathogenesis of SLE [89]. Several experiments have revealed that exo-
ogy in SLE is only in the early exploration stage. For the first time, our somes can also serve as nanocarriers and biomarkers of SLE [88]. Sun
group applied RBC membrane-camouflaged nanoparticles to treat lupus et al. used exosomes as a transport vehicle to deliver curcumin to target
mice [85]. Due to the large variability in blood concentrations of CsA, we the inflammatory cells [90]. This approach holds promise for use in SLE.
constructed RBC-coated and CsA-loaded PLGA nanoparticles (CsA-RNPs) The properties of exosomes alone are not enough to ensure the enrich-
to optimize the pharmacokinetic properties of CsA (Fig. 5a). According to ment of therapeutic agents in disease tissues, so exosomes are usually
the whole blood drug concentration curve, the half-life of CsA-RNPs is engineered to improve targeting ability [91]. Fang et al. constructed
2.05 times longer than that of uncoated nanoparticles. After CsA-RNPs engineered NIH 3T3 cell membrane nanovesicles expressing CD40 (CD40
treatment, the lupus serological indicators and organ damage have been NVs) to target CD40/CD40L axis to block B cell signal activation
effectively improved [85]. Guo et al. pioneered the use of cancer cell (Fig. 5d). It was found that MMF-loaded CD40 NVs inhibited B cell
membrane-coated nanoparticles named IM-MNPs to treat lupus mice activation and alleviated the disease progression in MRL/lpr mice [92].
(Fig. 5b). The 4T1 cancer cell was engineered with major histocompati- One problem with MSCs transplantation is the difficulty of cell storage
bility complex class I (MHC I) deficient to lower immunogenicity. After and possible tumor induction. Fortunately, some studies have shown that
incubation with INF-γ, the expression of CD155 and PD-L1 was upregu- exosomes from MSCs have similar immunomodulatory functions as
lated in 4T1 cells, which could increase the targeting of nanoparticles to MSCs. Therefore, exosomes with high stability and easy long-term stor-
CD4 T cells. The results revealed that dexamethasone-loaded IM-MNPs age are promising as cell-free therapy for SLE [78]. Due to the excellent
shaped the effector T cell/Treg balance and attenuated the progression of properties of EVs, lupus-related researchers should pay more attention to
SLE [86]. applying EVs and engineered EVs to SLE treatment.

Fig. 5. (a) Diagrammatic overview of the fabrication and application of CsA-RNPs. Reproduced with permission from Ref. [85], Copyright 2022 The Authors. (b)
Schematic of the preparation process and application of dexamethasone-loaded IM-MNPs. Reproduced with permission from Ref. [86] Copyright 2022 Elsevier Ltd. (c)
Immunoregulatory functions of EVs. Reproduced with permission from Ref. [93] Copyright 2022 Springer Nature Limited. (d) Schematic overview of MMF-loaded
CD40 NVs for SLE treatment. Reproduced with permission from Ref. [92] Copyright 2023 Wiley-VCH GmbH.

7
X. Hao et al. Biomedical Technology 4 (2023) 1–10

4. Conclusion and perspectives Foundation of China (82102511), the Natural Science Foundation of
Jiangsu (BK20210021), the Research Project of Jiangsu Province Health
SLE is a complicated autoimmune disease in which nearly all crucial Committee (M2021031), the National Natural Science Foundation of
elements of the immune system are involved in the underlying patho- China (82101184), Shenzhen Fundamental Research Program
genic mechanism. From the antimalarial drugs approved for lupus in the (JCYJ20210324102809024), Shenzhen PhD Start-up Program
1950s to the present, we have a more complete and in-depth under- (RCBS20210609103713045).
standing of lupus pathogenesis and management. However, it was not
until 2011 that belimumab was approved by the FDA for the treatment of References
lupus, filling the dilemma of no new drug available for nearly 60 years.
Rapid advances in engineering technology bring new opportunities for [1] A. Psarras, M. Wittmann, E.M. Vital, Emerging concepts of type I interferons in SLE
pathogenesis and therapy, Nat. Rev. Rheumatol. 18 (2022) 575–590.
lupus treatment. Compared with traditional therapeutic methods, nano- [2] R.A. Furie, R.F. van Vollenhoven, K. Kalunian, S. Navarra, J. Romero-Diaz,
carriers have multiple advantages in agents delivery for the treatment of V.P. Werth, X. Huang, G. Clark, H. Carroll, A. Meyers, et al., Trial of anti-BDCA2
SLE, such as enhanced drug solubility, sustained release, and passive antibody litifilimab for systemic lupus erythematosus, N. Engl. J. Med. 387 (2022)
894–904.
accumulation at inflammatory sites. When the surface of nanocarriers is [3] J. Tian, D. Zhang, X. Yao, Y. Huang, Q. Lu, Global epidemiology of systemic lupus
engineered, active targeting of tissues or cells in need of delivery can also erythematosus: a comprehensive systematic analysis and modelling study, Ann.
be achieved. Nanocarriers currently in lupus research are mainly used for Rheum. Dis. 82 (3) (2023 Mar) 351–356.
[4] M.R.W. Barber, C. Drenkard, T. Falasinnu, A. Hoi, A. Mak, N.Y. Kow,
the selective delivery of glucocorticoids and immunosuppressants. The E. Svenungsson, J. Peterson, A.E. Clarke, R. Ramsey-Goldman, Global epidemiology
work of Xu et al. provides another idea to improve the clearance of self- of systemic lupus erythematosus, Nat. Rev. Rheumatol. 17 (2021) 515–532.
antigens by apoptotic cells for treating lupus with nanocarriers [61]. [5] Systemic lupus erythematosus, Ann. Intern. Med. 172 (2020) ITC81–ITC96.
[6] F. Basta, F. Fasola, K. Triantafyllias, A. Schwarting, Systemic lupus erythematosus
Despite the excellent performance, it cannot be ignored that the
(SLE) therapy: the old and the new, Rheumatology and Therapy 7 (2020) 433–446.
biocompatibility and efficacy of nanocarriers in the human body still [7] A. Prete, I. Bancos, Glucocorticoid induced adrenal insufficiency, BMJ 374 (2021)
need to be further explored. CAR-T cell therapy has shown encouraging n1380.
achievements in both preclinical and clinical studies in refractory SLE. [8] C.B. Burness, P.L. McCormack, Belimumab, Drugs 71 (2011) 2435–2444.
[9] X. Yu, N. Chen, J. Xue, C.C. Mok, S.-C. Bae, X. Peng, W. Chen, H. Ren, X. Li,
The effect of CAT-T cell therapy lasted for a long time, and the patient's K. Noppakun, et al., Efficacy and safety of belimumab in patients with lupus
SLEDAI almost returned to 0. However, the largest sample in the study of nephritis: subgroup analyses of a phase 3 randomized trial in the east asian
CAR-T treatment of lupus is only 5 patients, and further research is population, Am J Kidney Dis. (2022 Sep 2). S0272-6386(22)00860-5.
[10] H.I. Brunner, C. Abud-Mendoza, D.O. Viola, I. Calvo Penades, D. Levy, J. Anton,
needed on larger samples and whether it can treat early or mild SLE. J.E. Calderon, V.G. Chasnyk, M.A. Ferrandiz, V. Keltsev, et al., Safety and efficacy of
Engineered MSCs are expected to solve the problem that cell trans- intravenous belimumab in children with systemic lupus erythematosus: results from
plantation in some patients is ineffective partially because of the strong a randomised, placebo-controlled trial, Ann. Rheum. Dis. 79 (2020) 1340–1348.
[11] N. Sch€afer, S. Gr€assel, New refinements aim to optimize articular cartilage tissue
immunomodulatory plasticity of MSCs. Microparticles and microgels engineering, Nat. Rev. Rheumatol. 19 (2023) 66–67.
encapsulating MSCs also provide intraperitoneal and subcutaneous [12] Y. Zhu, L. Cai, H. Chen, Y. Zhao, Developing organs-on-chips for biomedicine, Sci.
routes for MSCs transplantation. However, these studies on engineered Bull. 67 (2022) 1108–1111.
[13] L. Shang, Y. Zhao, W. Cui, Regenerative medicine entering a new era, Small 18
MSCs are still at the stage of animal models. CM-coated nanoparticles and (2022), 2204625.
EVs are currently considered as next-generation drug delivery platforms. [14] M. Zheng, H. Jia, H. Wang, L. Liu, Z. He, Z. Zhang, W. Yang, L. Gao, X. Gao, F. Gao,
The efficacy of nanoparticles encapsulated in RBC membrane and tumor Application of nanomaterials in the treatment of rheumatoid arthritis, RSC Adv. 11
(2021) 7129–7137.
CM has been effectively tested in MRL/lpr mice, but other CM could also
[15] R. He, L. Li, T. Zhang, X. Ding, Y. Xing, S. Zhu, Z. Gu, H. Hu, Recent advances of
be introduced to treat lupus. For example, neutrophil membrane-coated nanotechnology application in autoimmune diseases – a bibliometric analysis, Nano
nanoparticles can target sites of inflammation, which has been shown to Today 48 (2023), 101694.
work well in mice with rheumatoid arthritis. EVs themselves can be used [16] B. Ozbakir, B.J. Crielaard, J.M. Metselaar, G. Storm, T. Lammers, Liposomal
corticosteroids for the treatment of inflammatory disorders and cancer, J. Contr.
as therapeutic agents or as vehicles for the delivery of drugs or nucleic Release 190 (2014) 624–636.
acids. Therefore, more research on engineered EVs should focus on [17] D. Peer, J.M. Karp, S. Hong, O.C. Farokhzad, R. Margalit, R. Langer, Nanocarriers as
treating SLE. In general, engineering technology is a significant oppor- an emerging platform for cancer therapy, Nat. Nanotechnol. 2 (2007) 751–760.
[18] R.H. Fang, W. Gao, L. Zhang, Targeting drugs to tumours using cell membrane-
tunity to break the dilemma of no new drugs available for SLE. coated nanoparticles, Nat. Rev. Clin. Oncol. 20 (2023) 33–48.
[19] X. Jin, Y. Han, J.Q. Wang, L. Lu, CAR-T cell therapy: new hope for systemic lupus
CRediT authorship contribution statement erythematosus patients, Cell. Mol. Immunol. 18 (2021) 2581–2582.
[20] H. Zhu, B. Kong, M. Nie, C. Zhao, R. Liu, Y. Xie, Y. Zhao, L. Sun, ECM-inspired
peptide dendrimer microgels with human MSCs encapsulation for systemic lupus
Xubin Hao: Conceptualization, Writing – original draft. Yuze Wang: erythematosus treatment, Nano Today 43 (2022).
Writing – review & editing. Rui Liu: Writing – review & editing. Dagan [21] M. Nie, G. Chen, C. Zhao, J. Gan, M. Alip, Y. Zhao, L. Sun, Bio-inspired adhesive
porous particles with human MSCs encapsulation for systemic lupus erythematosus
Zhang: Funding acquisition, Writing – review & editing. Bin Kong: treatment, Bioact. Mater. 6 (2021) 84–90.
Funding acquisition, Writing – review & editing. Jun Liang: Conceptu- [22] A. Fanouriakis, N. Tziolos, G. Bertsias, D.T. Boumpas, Update οn the diagnosis and
alization, Funding acquisition, Writing – review & editing. Lingyun Sun: management of systemic lupus erythematosus, Ann. Rheum. Dis. 80 (2021) 14–25.
[23] R.S. Hardy, K. Raza, M.S. Cooper, Therapeutic glucocorticoids: mechanisms of
Supervision.
actions in rheumatic diseases, Nat. Rev. Rheumatol. 16 (2020) 133–144.
[24] D. Apostolopoulos, E.F. Morand, It hasn't gone away: the problem of glucocorticoid
Declaration of competing interest use in lupus remains, Rheumatology 56 (2017) i114–i122.
[25] N. Acharya, Glucocorticoid withdrawal in lupus - to do or not to do? Ann. Rheum.
Dis. 81 (2022) e44.
The authors declare that they have no known competing financial [26] W. Xiong, R.G. Lahita, Pragmatic approaches to therapy for systemic lupus
interests or personal relationships that could have appeared to influence erythematosus, Nat. Rev. Rheumatol. 10 (2014) 97–107.
the work reported in this paper. [27] A.M. Schjerning, P. McGettigan, G. Gislason, Cardiovascular effects and safety of
(non-aspirin) NSAIDs, Nat. Rev. Cardiol. 17 (2020) 574–584.
[28] M.J. Domper Arnal, G. Hijos-Mallada, A. Lanas, Gastrointestinal and cardiovascular
Acknowledgments adverse events associated with NSAIDs, Expet Opin. Drug Saf. 21 (2022) 373–384.
[29] O. Araújo, J. Hernandez-Rodríguez, L. Pelegrín, M. Feliu, M. Boland, H. Hernandez-
Negrín, A. Adan, G. Espinosa, R. Cervera, Why lupus patients discontinue
This work was supported by the fundings for Clinical Trials from the antimalarials in real life: a 50 years-experience from a reference centre, Lupus 31
Affiliated Drum Tower Hospital, Medical School of Nanjing University (2022) 1344–1354.
(2022-LCYJ-MS-38), the Puai Medical Research Fund of Beijing Bethune [30] S.J. Lee, E. Silverman, J.M. Bargman, The role of antimalarial agents in the
treatment of SLE and lupus nephritis, Nat. Rev. Nephrol. 7 (2011) 718–729.
Charitable Foundation (PAYJ-008), the National Natural Science

8
X. Hao et al. Biomedical Technology 4 (2023) 1–10

[31] G. Ruiz-Irastorza, D. Martin-Iglesias, A. Soto-Peleteiro, Update on antimalarials and [61] N. Xu, J. Li, Y. Gao, N. Zhou, Q. Ma, M. Wu, Y. Zhang, X. Sun, J. Xie, G. Shen, et al.,
systemic lupus erythematosus, Curr. Opin. Rheumatol. 32 (2020) 572–582. Apoptotic cell-mimicking gold nanocages loaded with LXR agonist for attenuating
[32] G. Figueroa-Parra, C.M. Gamboa-Alonso, A.L. De-Leon-Ibarra, D.A. Galarza- the progression of murine systemic lupus erythematosus, Biomaterials 197 (2019)
Delgado, Update of the EULAR recommendations for the management of SLE: don't 380–392.
forget chloroquine, Ann. Rheum. Dis. 79 (2019) e114, 2020. [62] S.G. Tangye, C.S. Ma, R. Brink, E.K. Deenick, The good, the bad and the ugly - TFH
[33] D.J. Wallace, V.S. Gudsoorkar, M.H. Weisman, S.R. Venuturupalli, New insights into cells in human health and disease, Nat. Rev. Immunol. 13 (2013) 412–426.
mechanisms of therapeutic effects of antimalarial agents in SLE, Nat. Rev. [63] J. Zhang, Q. Guo, D. Dai, J. Yu, L. Wang, Z. Wu, H. Ding, N. Shen, Y. Duan,
Rheumatol. 8 (2012) 522–533. Rapamycin-encapsulated costimulatory ICOS/CD40L-bispecific nanoparticles
[34] D.H. Wang, D.J. Wallace, New insights into systemic lupus erythematosus therapies: restrict pathogenic helper T-B-cell interactions while in situ suppressing mTOR for
2010-2020, J. Clin. Rheumatol. 28 (2022) e217–e221. lupus treatment, Biomaterials 289 (2022), 121766.
[35] N. Schwartz, A.D. Stock, C. Putterman, Neuropsychiatric lupus: new mechanistic [64] M.A. Grimaudo, A. Concheiro, C. Alvarez-Lorenzo, Nanogels for regenerative
insights and future treatment directions, Nat. Rev. Rheumatol. 15 (2019) 137–152. medicine, J. Contr. Release 313 (2019) 148–160.
[36] A. Mohamed, Y. Chen, H. Wu, J. Liao, B. Cheng, Q. Lu, Therapeutic advances in the [65] M. Look, E. Stern, Q.A. Wang, L.D. DiPlacido, M. Kashgarian, J. Craft, T.M. Fahmy,
treatment of SLE, Int. Immunopharm. 72 (2019) 218–223. Nanogel-based delivery of mycophenolic acid ameliorates systemic lupus
[37] Y.A. Heo, Voclosporin: first approval, Drugs 81 (2021) 605–610. erythematosus in mice, J. Clin. Investig. 123 (2013) 1741–1749.
[38] M. Kostopoulou, A. Fanouriakis, G. Bertsias, D.T. Boumpas, Treatment of lupus: [66] J. Liu, X. Zhang, X. Cao, Dendritic cells in systemic lupus erythematosus: from
more options after a long wait, Ann. Rheum. Dis. 81 (2022) 753–756. pathogenesis to therapeutic applications, J. Autoimmun. 132 (2022), 102856.
[39] E.D. Deeks, Anifrolumab: first approval, Drugs 81 (2021) 1795–1802. [67] M. Look, W.M. Saltzman, J. Craft, T.M. Fahmy, The nanomaterial-dependent
[40] A. Mohamed, Y. Chen, H. Wu, J. Liao, B. Cheng, Q. Lu, Therapeutic advances in the modulation of dendritic cells and its potential influence on therapeutic
treatment of SLE, Int. Immunopharm. 72 (2019) 218–223. immunosuppression in lupus, Biomaterials 35 (2014) 1089–1095.
[41] Y. Shao, Z. Yang, Progress in polymer single-chain based hybrid nanoparticles, [68] R. Phillips, CAR T cells induce drug-free SLE remission, Nat. Rev. Rheumatol. 18
Prog. Polym. Sci. 133 (2022), 101593. (2022), 671-671.
[42] D. Rostamzadeh, S.R. Razavi, S. Esmaeili, S. Dolati, M. Ahmahi, S. Sadreddini, [69] A. Mullard, FDA approves first CAR T therapy, Nat. Rev. Drug Discov. 16 (2017),
F. Jadidi-Niaragh, M. Yousefi, Application of nanoparticle technology in the 669-669.
treatment of Systemic lupus erythematous, Biomed. Pharmacother. 83 (2016) [70] X. Jin, Q. Xu, C. Pu, K. Zhu, C. Lu, Y. Jiang, L. Xiao, Y. Han, L. Lu, Therapeutic
1154–1163. efficacy of anti-CD19 CAR-T cells in a mouse model of systemic lupus
[43] C.B. Chighizola, V.H. Ong, P.L. Meroni, The use of cyclosporine A in rheumatology: erythematosus, Cell. Mol. Immunol. 18 (2021) 1896–1903.
a 2016 comprehensive review, Clin. Rev. Allergy Immunol. 52 (2017) 401–423. [71] R. Kansal, N. Richardson, I. Neeli, S. Khawaja, D. Chamberlain, M. Ghani, Q-u-
[44] R. Ganugula, M. Arora, D. Zou, S.K. Agarwal, C. Mohan, M.N.V.R. Kumar, A highly a Ghani, L. Balazs, S. Beranova-Giorgianni, F. Giorgianni, et al., Sustained B cell
potent lymphatic system-targeting nanoparticle cyclosporine prevents depletion by CD19-targeted CAR T cells is a highly effective treatment for murine
glomerulonephritis in mouse model of lupus, Sci. Adv. 6 (2020), eabb3900. lupus, Sci. Transl. Med. 11 (2019), eaav1648.
[45] B. Jiang, Y. Zhang, Y. Li, Y. Chen, S. Sha, L. Zhao, D. Li, J. Wen, J. Lan, Y. Lou, et al., [72] D. Mougiakakos, G. Kr€ onke, S. V€
olkl, S. Kretschmann, M. Aigner, S. Kharboutli,
A tissue-tended mycophenolate-modified nanoparticle alleviates systemic lupus S. B€oltz, B. Manger, A. Mackensen, G. Schett, CD19-Targeted CAR T cells in
erythematosus in MRL/lpr mouse model mainly by promoting local M2-like refractory systemic lupus erythematosus, N. Engl. J. Med. 385 (2021) 567–569.
macrophagocytes polarization, Int. J. Nanomed. 17 (2022) 3251–3267. [73] W. Zhang, J. Feng, A. Cinquina, Q. Wang, H. Xu, Q. Zhang, L. Sun, Q. Chen, L. Xu,
[46] J. Hu, M. Wang, X. Xiao, B. Zhang, Q. Xie, X. Xu, S. Li, Z. Zheng, D. Wei, X. Zhang, K. Pinz, et al., Treatment of systemic lupus erythematosus using BCMA-CD19
A novel long-acting azathioprine polyhydroxyalkanoate nanoparticle enhances compound CAR, Stem Cell Rev Rep 17 (2021) 2120–2123.
treatment efficacy for systemic lupus erythematosus with reduced side effects, [74] A. Mackensen, F. Muller, D. Mougiakakos, S. Boltz, A. Wilhelm, M. Aigner, S. Volkl,
Nanoscale 12 (2020) 10799–10808. D. Simon, A. Kleyer, L. Munoz, et al., Anti-CD19 CAR T cell therapy for refractory
[47] H. Shimizu, Y. Hori, S. Kaname, K. Yamada, N. Nishiyama, S. Matsumoto, K. Miyata, systemic lupus erythematosus, Nat. Med. 28 (2022) 2124–2132.
M. Oba, A. Yamada, K. Kataoka, T. Fujita, siRNA-based therapy ameliorates [75] R. Khatri, T. Linn, Pancreatic mesenchymal stem cells might protect β-cells, Nat.
glomerulonephritis, J. Am. Soc. Nephrol. 21 (2010) 622–633. Rev. Endocrinol. 18 (2022) 725–726.
[48] W. Pan, S. Zhu, D. Dai, Z. Liu, D. Li, B. Li, N. Gagliani, Y. Zheng, Y. Tang, [76] D. Wang, H. Zhang, J. Liang, H. Wang, B. Hua, X. Feng, G.S. Gilkeson, D. Farge,
M.T. Weirauch, et al., MiR-125a targets effector programs to stabilize Treg- S. Shi, L. Sun, A long-term follow-up study of allogeneic mesenchymal stem/stromal
mediated immune homeostasis, Nat. Commun. 6 (2015) 7096. cell transplantation in patients with drug-resistant systemic lupus erythematosus,
[49] J. Zhang, C. Chen, H. Fu, J. Yu, Y. Sun, H. Huang, Y. Tang, N. Shen, Y. Duan, Stem Cell Rep. 10 (2018) 933–941.
MicroRNA-125a-Loaded polymeric nanoparticles alleviate systemic lupus [77] J. Liang, H. Zhang, B. Hua, H. Wang, L. Lu, S. Shi, Y. Hou, X. Zeng, G.S. Gilkeson,
erythematosus by restoring effector/regulatory T cells balance, ACS Nano 14 (2020) L. Sun, Allogenic mesenchymal stem cells transplantation in refractory systemic
4414–4429. lupus erythematosus: a pilot clinical study, Ann. Rheum. Dis. 69 (2010) 1423–1429.
[50] Y. Lu, Z. Yue, J. Xie, W. Wang, H. Zhu, E. Zhang, Z. Cao, Micelles with ultralow [78] A. Li, F. Guo, Q. Pan, S. Chen, J. Chen, H.F. Liu, Q. Pan, Mesenchymal stem cell
critical micelle concentration as carriers for drug delivery, Nat Biomed Eng 2 (2018) therapy: hope for patients with systemic lupus erythematosus, Front. Immunol. 12
318–325. (2021), 728190.
[51] X. Clemente-Casares, P. Santamaria, Nanomedicine in autoimmunity, Immunol. [79] N. Che, X. Li, L. Zhang, R. Liu, H. Chen, X. Gao, S. Shi, W. Chen, L. Sun, Impaired B
Lett. 158 (2014) 167–174. cell inhibition by lupus bone marrow mesenchymal stem cells is caused by reduced
[52] Z. Jia, X. Wang, X. Wei, G. Zhao, K.W. Foster, F. Qiu, Y. Gao, F. Yuan, F. Yu, CCL2 expression, J. Immunol. 193 (2014) 5306–5314.
G.M. Thiele, et al., Micelle-forming dexamethasone prodrug attenuates nephritis in [80] J. Xu, J. Chen, W. Li, W. Lian, J. Huang, B. Lai, L. Li, Z. Huang, Additive therapeutic
lupus-prone mice without apparent glucocorticoid side effects, ACS Nano 12 (2018) effects of mesenchymal stem cells and IL-37 for systemic lupus erythematosus,
7663–7681. J. Am. Soc. Nephrol. 31 (2020) 54–65.
[53] Z. Zhao, X. Xu, H. Jiang, K.W. Foster, Z. Jia, X. Wei, N. Chen, S.R. Goldring, [81] M. Desgres, P. Menasche, Clinical translation of pluripotent stem cell therapies:
M.K. Crow, D. Wang, Preclinical dose-escalation study of ZSJ-0228, a polymeric challenges and considerations, Cell Stem Cell 25 (2019) 594–606.
dexamethasone prodrug, in the treatment of murine lupus nephritis, Mol. Pharm. 18 [82] J. Galipeau, L. Sensebe, Mesenchymal stromal cells: clinical challenges and
(2021) 4188–4197. therapeutic opportunities, Cell Stem Cell 22 (2018) 824–833.
[54] N. Grimaldi, F. Andrade, N. Segovia, L. Ferrer-Tasies, S. Sala, J. Veciana, N. Ventosa, [83] A.S. Abu Lila, H. Kiwada, T. Ishida, The accelerated blood clearance (ABC)
Lipid-based nanovesicles for nanomedicine, Chem. Soc. Rev. 45 (2016) 6520–6545. phenomenon: clinical challenge and approaches to manage, J. Contr. Release 172
[55] Y. Barenholz, Doxil® — the first FDA-approved nano-drug: lessons learned, (2013) 38–47.
J. Contr. Release 160 (2012) 117–134. [84] C.M. Hu, L. Zhang, S. Aryal, C. Cheung, R.H. Fang, L. Zhang, Erythrocyte
[56] Y. Scindia, U. Deshmukh, P.R. Thimmalapura, H. Bagavant, Anti-alpha8 integrin membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform,
immunoliposomes in glomeruli of lupus-susceptible mice: a novel system for Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 10980–10985.
delivery of therapeutic agents to the renal glomerulus in systemic lupus [85] X. Hao, H. Zhang, R. Liu, J. Che, D. Zhang, J. Liang, L. Sun, Red blood cell
erythematosus, Arthritis Rheum. 58 (2008) 3884–3891. membrane functionalized biomimetic nanoparticles for systemic lupus
[57] E. Moallem, E. Koren, R. Ulmansky, G. Pizov, M. Barlev, Y. Barenholz, Y. Naparstek, erythematosus treatment, Materials Today Advances 16 (2022).
A liposomal steroid nano-drug for treating systemic lupus erythematosus, Lupus 25 [86] Q. Guo, C. Chen, Z. Wu, W. Zhang, L. Wang, J. Yu, L. Li, J. Zhang, Y. Duan,
(2016) 1209–1216. Engineered PD-1/TIGIT dual-activating cell-membrane nanoparticles with
[58] X. Guo, L. Wang, K. Duval, J. Fan, S. Zhou, Z. Chen, Dimeric drug polymeric dexamethasone act synergistically to shape the effector T cell/Treg balance and
micelles with acid-active tumor targeting and FRET-traceable drug release, Adv. alleviate systemic lupus erythematosus, Biomaterials 285 (2022), 121517.
Mater. 30 (2018). [87] I.K. Herrmann, M.J.A. Wood, G. Fuhrmann, Extracellular vesicles as a next-
[59] H. Zhang, R. Fu, C. Guo, Y. Huang, H. Wang, S. Wang, J. Zhao, N. Yang, Anti-dsDNA generation drug delivery platform, Nat. Nanotechnol. 16 (2021) 748–759.
antibodies bind to TLR4 and activate NLRP3 inflammasome in lupus monocytes/ [88] Y. Fang, J. Ni, Y.S. Wang, Y. Zhao, L.Q. Jiang, C. Chen, R.D. Zhang, X. Fang,
macrophages, J. Transl. Med. 14 (2016) 156. P. Wang, H.F. Pan, Exosomes as biomarkers and therapeutic delivery for
[60] L. Diao, J. Tao, Y. Wang, Y. Hu, W. He, Co-delivery of dihydroartemisinin and autoimmune diseases: opportunities and challenges, Autoimmun. Rev. 22 (2022),
HMGB1 siRNA by TAT-modified cationic liposomes through the TLR4 signaling 103260.
pathway for treatment of lupus nephritis, Int. J. Nanomed. 14 (2019) 8627–8645.

9
X. Hao et al. Biomedical Technology 4 (2023) 1–10

[89] Y. Fei, Q. Liu, N. Peng, G. Yang, Z. Shen, P. Hong, S. Wang, K. Rui, D. Cui, Exosomes [91] A. Ortega, O. Martinez-Arroyo, M.J. Forner, R. Cortes, Exosomes as drug delivery
as crucial players in pathogenesis of systemic lupus erythematosus, Journal of systems: endogenous nanovehicles for treatment of systemic lupus erythematosus,
Immunology Research 2022 (2022), 8286498. Pharmaceutics 13 (2021) 3.
[90] D. Sun, X. Zhuang, X. Xiang, Y. Liu, S. Zhang, C. Liu, S. Barnes, W. Grizzle, D. Miller, [92] T. Fang, B. Li, M. Li, Y. Zhang, Z. Jing, Y. Li, T. Xue, Z. Zhang, W. Fang, Z. Lin, et al.,
H.G. Zhang, A novel nanoparticle drug delivery system: the anti-inflammatory Engineered cell membrane vesicles expressing CD40 alleviate system lupus
activity of curcumin is enhanced when encapsulated in exosomes, Mol. Ther. 18 nephritis by intervening B cell activation, Small Methods (2023), e2200925.
(2010) 1606–1614. [93] E.I. Buzas, The roles of extracellular vesicles in the immune system, Nat. Rev.
Immunol. (2022) 1–15.

10

You might also like