Thanks to visit codestin.com
Credit goes to bmcbiol.biomedcentral.com

Skip to main content
BMC is moving to Springer Nature Link. Visit this journal in its new home.

Design and development of a bispecific antibody targeting BAFF and IL-17 for systemic lupus erythematosus treatment

Abstract

Background

This study aimed to develop and evaluate a bispecific single-chain variable fragment (bsscFv) targeting B-cell activating factor (BAFF) and interleukin-17 (IL-17) for the treatment of systemic lupus erythematosus (SLE).

Methods

The bsscFv was engineered by linking single-chain variable fragments (scFvs) specific for BAFF and IL-17 with a flexible peptide linker. It was expressed in E. coli BL21 and purified using affinity chromatography. Binding affinities to BAFF and IL-17 were assessed by enzyme-linked immunosorbent assay (ELISA). In vitro neutralization assays were conducted using Raji and HT-29 cell cultures. In vivo therapeutic efficacy was evaluated in an MRL/lpr mouse model of SLE, with 10 mice per group. Statistical significance was determined using a Student’s t-test for comparison of two groups.

Results

The bsscFv demonstrated strong binding to both BAFF and IL-17 in ELISA. In vitro, it inhibited BAFF-induced B-cell survival, proliferation, and immunoglobulin production, as well as IL-17-induced inflammatory cytokine secretion in HT-29 cells. In the MRL/lpr mouse model, bsscFv treatment significantly reduced autoantibody levels (p < 0.05), proteinuria, renal pathology, and cytokine expression in a dose-dependent manner compared to controls.

Conclusions

The bsscFv exhibited potent neutralizing activity in vitro and therapeutic efficacy in vivo, suggesting it as a promising bispecific therapeutic agent for the treatment of SLE. Further studies are warranted to explore its clinical potential.

Background

Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder characterized by the production of autoantibodies, immune complex deposition, and multi-organ damage [1, 2]. The pathogenesis of SLE involves a complex interplay of genetic, environmental, and immunological factors, leading to the breakdown of self-tolerance and the development of autoimmunity [3, 4]. Despite advances in our understanding of SLE pathogenesis and the availability of various treatment options, many patients continue to experience significant morbidity and mortality, highlighting the need for novel therapeutic strategies [5].

B-cell activating factor (BAFF) and interleukin-17 (IL-17) are two key cytokines implicated in the pathogenesis of SLE [6,7,8]. BAFF, a member of the tumor necrosis factor (TNF) superfamily, plays a crucial role in B-cell survival, maturation, and autoantibody production [9, 10]. Elevated levels of BAFF have been observed in SLE patients and correlate with disease activity and autoantibody titers [11]. IL-17, produced by T helper 17 (Th17) cells, has been shown to contribute to SLE pathogenesis by promoting inflammation, tissue damage, and the recruitment of immune cells [12, 13]. Increased levels of IL-17 have been detected in the serum and target organs of SLE patients, suggesting its involvement in disease progression [14, 15].

Given the central roles of BAFF and IL-17 in SLE pathogenesis, targeting these cytokines represents a promising therapeutic approach. Monoclonal antibodies targeting BAFF, such as belimumab, have demonstrated clinical efficacy in SLE patients, leading to their approval for the treatment of SLE [16, 17]. Similarly, IL-17 inhibitors have shown potential in preclinical models of SLE and are currently being investigated in clinical trials [18, 19].

Bispecific antibodies, engineered to recognize two different antigens, have emerged as a powerful tool for the treatment of various diseases, including autoimmune disorders [20, 21]. By simultaneously targeting multiple disease-related pathways, bispecific antibodies offer the potential for enhanced therapeutic efficacy and reduced side effects compared to conventional monoclonal antibodies [22].

In this study, we aimed to develop and evaluate a novel bispecific single-chain variable fragment (bsscFv) targeting both BAFF and IL-17 for the treatment of SLE. We hypothesized that the simultaneous inhibition of these two key cytokines would result in enhanced therapeutic efficacy compared to targeting either cytokine alone. To test this hypothesis, we generated a panel of bsscFv using phage display technology and assessed their binding affinity, specificity, and in vitro functional activity. The lead bsscFv was then evaluated in a well-established murine model of SLE to determine its efficacy in reducing autoantibody production, kidney damage, and systemic inflammation. Our results demonstrate the potential of bsscFvs targeting BAFF and IL-17 as a novel therapeutic strategy for the treatment of SLE.

Results

Protein expression and purification

Recombinant human BAFF and IL-17 proteins were expressed in HEK293F cells and purified via Ni–NTA affinity chromatography. SDS-PAGE showed bands at the expected molecular weights of 18 kDa for BAFF and 16 kDa for IL-17 (Additional file 1: Fig. S1A). The purity of the proteins was > 95%, confirmed by densitometric analysis of Coomassie Blue-stained gels. Identity was further validated by Western blot using anti-His antibodies (Additional file 1: Fig. S1B).

Antibody titers and spleen cell isolation

Mice immunized with recombinant BAFF and IL-17 exhibited significantly higher antigen-specific antibody titers compared to pre-immune sera. Endpoint titers for anti-BAFF and anti-IL-17 antibodies reached 1:204,800 and 1:102,400, respectively, after the fourth immunization (Additional file 1: Fig. S1C and S1D). Spleens were harvested, and single-cell suspensions were prepared for antibody library construction, with cell viability exceeding 90% as assessed by trypan blue exclusion.

PCR of antibody chains and construction of scFv phage library

Total RNA was extracted from isolated spleen cells, and cDNA was synthesized using oligo(dT) primers. PCR amplification of the antibody light (VL) and heavy (VH) chain variable regions produced bands of approximately 400 bp and 500 bp, respectively (Additional file 1: Fig. S2A and S2B). The purified VL and VH fragments were cloned into the pCANTAB 5E vector, resulting in a phage library containing 10.5 × 1010 independent clones (Additional file 1: Table 2). PCR screening revealed > 93% of clones had inserts of the expected size (Additional file 1: Fig. S2C and S2D). Next-generation sequencing (NGS) analysis showed the library's VH and VK gene distribution matched natural germline frequencies (Additional file 1: Fig. S3A and S3B), with CDR3 sequence lengths following a normal distribution (Additional file 1: Fig. S3C) and random pairing of heavy and light chain variable regions (Additional file 1: Fig. S3D), indicating excellent sequence diversity for subsequent antibody screening.

Helper phage amplification and scFv library quality evaluation

TG1 plates infected with the helper phage were subjected to gradient dilution, and colony counts were used to calculate the titer of amplified M13K07 helper phage (Additional file 1: Table 3). Using a 10–6 dilution, 211 colonies were counted, resulting in a titer of 5.27 × 1014 pfu/mL from a total of 20 mL. This titer meets the requirements for subsequent screening. Similarly, the amplified scFv phage library was evaluated by gradient dilution of TG1 plates infected with the phage, yielding 125/98 colonies at a 10–3 dilution (Additional file 1: Table 4). The final titer of the phage library was 6.25 × 1012/4.9 × 1012 pfu/mL, indicating sufficient quantity for further screening.

Phage display selection

The scFv library underwent five rounds of phage display selection against recombinant BAFF and IL-17 proteins (Additional file 1: Table 5). In each round, phages binding to the targets were eluted and amplified. After the fifth round, individual clones were screened using phage ELISA, and positive clones (OD450nm > 2) were sequenced (Additional file 1: Fig. S4). Two unique scFv clones, scFv1 and scFv2, were identified as specific binders to BAFF and IL-17, respectively.

Construction and expression of bsscFv

As shown in Fig. 1A, scFv1 and scFv2 sequences were fused via a flexible linker (G4S)2 to create the bsscFv construct. The bsscFv gene was cloned into a pET28a(+) vector and transfected into BL21(DE3) cells for protein expression (Additional file 1: Fig. S5A). The protein was purified using affinity chromatography from disrupted cell lysates. SDS-PAGE and Western blotting with anti-His antibodies confirmed the purified protein (Fig. 1B, C). To quantitatively assess the binding affinity of the bispecific antibody, we performed label-free kinetic measurements using the Gator Prime system. The determined kinetic parameters showed that bsscFv exhibited high-affinity binding to both BAFF and IL-17, with KD values in the low nanomolar range (Table 1). Importantly, pre-saturation of bsscFv with either BAFF or IL-17 did not significantly alter the binding kinetics to the second target, indicating independent, non-competitive engagement of both epitopes (Additional file 1: Fig. S5B, S5C). These results confirm that the bispecific antibody maintains dual specificity and high-affinity binding to each target independently, even in the presence of the other.

Fig. 1
figure 1

Schematic of bispecific antibody construction and mechanism of action. A Schematic representation of bispecific single-chain antibody construction. B Purification and expression of single-chain antibodies. M represents the protein marker; bsscFv indicates the expression and purification of the bispecific single-chain antibody; scFv1 and scFv2 represent the anti-BAFF and anti-IL-17 single-chain antibodies, respectively. C Western blot (WB) identification of single-chain antibodies. The primary antibody is mouse anti-His tag; the secondary antibody is HRP-conjugated goat anti-mouse. D Schematic illustration of the mechanism of action of the bispecific antibody

Table 1 Kinetic parameters of bsscFv and monospecific scFvs binding to BAFF and IL-17

In vitro experiments

The neutralizing activity of bsscFv was assessed in Raji and HT-29 cells. BAFF promoted Raji cell proliferation (Fig. 2A), and IL-17 induced CXCL1 secretion in HT-29 cells (Fig. 2B). bsscFv inhibited BAFF-induced Raji cell proliferation and IL-17-mediated CXCL1 secretion. IL-17 did not affect bsscFv's inhibition of Raji cell proliferation (Fig. 2C), nor did BAFF affect its inhibition of CXCL1 secretion (Fig. 2D). The IC50 of bsscFv for Raji cell proliferation was 0.39 ± 0.04 nM, similar to scFv1 (0.43 ± 0.02 nM), and for HT-29 cells, it was 4.63 ± 0.35 nM, comparable to scFv2 (4.12 ± 0.19 nM). These results show that bsscFv blocks both BAFF and IL-17 effectively.

Fig. 2
figure 2

In vitro and in vivo functional validation of BAFF and IL-17. A BAFF protein promotes Raji cell proliferation in a dose-dependent manner. B IL-17 protein stimulates CXCL1 secretion in HT-29 cells. C Single-chain antibodies inhibit BAFF-induced cell proliferation. D Single-chain antibodies inhibit IL-17-induced CXCL1 secretion. E qPCR analysis of CD19 mRNA expression, a B lymphocyte surface marker, in mouse spleens. F Measurement of CXCL1 levels in mouse peripheral blood using a commercial ELISA kit. G Detection of bsscFv levels in mouse peripheral blood after intravenous injection of bsscFv using ELISA with different coating antigens (anti-G4S antibody, BAFF protein, IL-17 protein). H Serum concentrations of BAFF at indicated time points following intravenous administration of different doses of bsscFv in mice. I Serum concentrations of IL-17 at indicated time points following intravenous administration of different doses of bsscFv in mice. J Changes in B cell levels in mouse peripheral blood after intravenous injection of bsscFv

In vivo experiments

A single injection of scFv1/bsscFv significantly reduced CD19 transcription levels in mouse spleens (Fig. 2E), indicating effective BAFF neutralization. Similarly, scFv2/bsscFv injection significantly decreased CXCL1 levels in mice (Fig. 2F), demonstrating effective IL-17 antagonism.

PK/PD

The pharmacokinetic (PK) profile of bsscFv was assessed in mice following IV doses of 1, 5, or 10 mg/kg and SC doses of 5 mg/kg. The PK data showed non-linear behavior, with serum concentrations declining in a dose-dependent manner. The area under the curve (AUC0-∞) increased disproportionately with higher doses, and the terminal half-life extended from 3 to 8 days, likely due to target-mediated elimination via BAFF. SC bioavailability was 93%, with peak concentrations observed 3 days post-injection. Binding assays confirmed bsscFv's functional integrity and no antidrug antibodies (Fig. 2G). Both BAFF and IL-17 levels in serum were modulated in a dose-dependent manner following bsscFv administration, reflecting the pharmacodynamic (PD) effects of the antibody (Fig. 2H, I). PD analysis showed a dose-dependent reduction in circulating B cells, with 50% ~ 60% inhibition at 2 months post-5 mg/kg injection (Fig. 2J), confirming BAFF neutralization and bsscFv’s biological activity.

Effect of bsscFv treatment on urinary protein and autoantibody levels in MRL/lpr mice

Mice were divided into five groups: BALB/c control (n = 5), MRL/lpr model (n = 5), and MRL/lpr model with low (1 mg/kg, n = 5), medium (5 mg/kg, n = 5), or high (10 mg/kg, n = 5) doses of bsscFv, administered intraperitoneally twice weekly for 8 weeks. As shown in Fig. 3A-H, urinary protein levels decreased significantly in the medium- and high-dose groups. BUN levels also showed a dose-dependent reduction. Anti-dsDNA antibody levels, serum creatinine (Scr), ANA, anti-snRNP/Sm antibodies, and SLE-IgG levels were lower in the bsscFv-treated groups. The Urine Protein/Creatinine Ratio was significantly reduced in the medium- and high-dose groups.

Fig. 3
figure 3

Quantitative ELISA measurement of urinary protein and autoantibody levels in mice. A Urinary protein levels measured over 24 h. B Blood urea nitrogen (BUN) levels. C Anti-double-stranded DNA antibody levels. D Serum creatinine levels. E Antinuclear antibody (ANA) levels. F Anti-snRNP/Sm antibody levels. G Anti-SLE antibody levels. H Urinary protein/creatinine ratio. I qPCR analysis of SLE-related inflammatory factors in mouse kidneys. J Western blot analysis of SLE-related inflammatory factors in mouse kidneys. K Grayscale analysis of panel J using ImageJ. In panels A-H, the x-axis represents the following: 1, BALB/c control mice; 2, MRL/lpr model control mice; 3, 4, and 5, represent MRL/lpr model mice treated with low, medium, and high doses, respectively. In panel J, lane 1 represents the kidney of a BALB/c control mouse, lane 2 represents the kidney of an MRL/lpr model control mouse, and lane 3 represents the kidney of an MRL/lpr model mouse treated with a high dose

Influence of bsscFv treatment on cytokine levels and correlation with target distribution in MRL/lpr mice

As illustrated in Fig. 3I, expression levels of Il-2, Il-4, Il-6, Il-10, Ifnα, Tnfα and Cd83 in murine peripheral blood mononuclear cells were downregulated following bsscFv treatment. Figure 3J and K indicates that protein levels of IL-2, IL-4, IL-6, IL-10, IFN-α and TNF-α in kidney tissues were significantly reduced as a result of bsscFv treatment.

Histopathological improvements and reduced immune complex deposition in bsscFv-treated MRL/lpr mice

Figure 4A-C shows that HE, PAS, and Masson’s trichrome staining revealed improved renal pathology in the bsscFv-treated groups, with reduced glomerular and tubulointerstitial lesions. In HE staining, yellow arrows point to glomeruli, red arrows to renal tubules, and black arrows to interstitial areas. The MRL/lpr Control group exhibited tubular epithelial loosening, edema, and detachment. However, the treated group showed well-preserved, rounded tubular epithelial cells with orderly brush borders, confirming the treatment’s effectiveness. PAS and Masson’s trichrome staining further confirmed significant improvements in renal pathology in the treatment group. As shown in Fig. 4D and E, immune complex deposition in the kidneys was notably reduced in the bsscFv-treated groups, further supporting the therapeutic efficacy.

Fig. 4
figure 4

Immunohistochemical analysis of mouse kidneys. A Hematoxylin and eosin (HE) staining of mouse kidneys. Periodic acid-Schiff (PAS) staining of mouse kidneys. Masson’s trichrome staining of mouse kidneys. B Quantification of the proportion of PAS-positive area. C Quantification of the proportion of Masson-positive area. D Immunofluorescent staining of mouse kidney sections using FITC-conjugated goat anti-mouse IgG (H + L) to detect deposited immune complexes. Representative images from each group are shown. Quantitative analysis of fluorescence intensity and positive area is presented in panel (E)

Transcriptomic and metabolomic analyses reveal therapeutic effects of bsscFv treatment in MRL/lpr mice

Transcriptomic and metabolomic analyses of kidneys from the MRL/lpr model (M) and bispecific antibody-treated group (T) showed that bsscFv targeted immune responses and metabolic pathways. GO enrichment analysis revealed upregulation of immune-related processes, such as responses to stimuli, suggesting improved immune defense and reduced autoimmune damage. Enriched genes related to adaptive immunity and regulation indicated reduced pathological immune responses. Cellular components like phagocytic vesicles and immunoglobulin complexes suggested increased phagocytosis and antibody-mediated immunity, potentially alleviating kidney damage (Fig. 5A). KEGG pathway analysis highlighted metabolic pathways, such as peroxisome, phagosome, and fatty acid degradation, indicating reduced oxidative stress and inflammation. Lipid metabolism regulation, particularly unsaturated fatty acid biosynthesis, suggests protection from lipid peroxidation and kidney damage (Fig. 5B). Metabolomic clustering revealed higher levels of metabolites like (4-Oxo-1,2,3-benzotriazin-3(4H)-yl) acetic acid and anti-inflammatory compounds (Beclomethasone, Flurandrenolide) in the treated group (Fig. 5C). KEGG heatmap analysis showed changes in nucleotide and lipid metabolism, suggesting that bsscFv reduced metabolic imbalances and kidney damage associated with lipid disorders (Fig. 5D).

Fig. 5
figure 5

Transcriptomic and metabolomic analysis of mouse kidneys. A Dot plot of Gene Ontology (GO) enrichment analysis of differentially expressed transcripts. The horizontal axis shows the rich factor (the ratio of differentially expressed to all expressed transcripts in the GO term), and the vertical axis represents Gene Ontology terms. Dot color indicates p-value, and dot size reflects the number of differentially expressed transcripts. B Bar plot of KEGG pathway enrichment analysis for differentially expressed transcripts. The horizontal axis shows the number of differentially expressed transcripts, and the vertical axis represents KEGG pathways, with colors indicating pathway categories. C Heatmap of hierarchical clustering comparing group M and group T. The horizontal axis represents different groups, while the vertical axis shows differentially expressed metabolites. Red and blue colors indicate higher and lower expression, respectively. D KEGG heatmap comparing group M and group T. The horizontal axis represents samples, and the vertical axis shows KEGG metabolic pathways, with background color indicating pathway categories. The color blocks represent relative abundance of metabolites, with darker blue indicating higher abundance

Analysis of antigen–antibody binding sites

The three-dimensional structure of bsscFv was predicted using AlphaFold (Additional file 1: Fig. S6A). The structures of BAFF (PDB ID: 1KD7) and IL-17 (PDB ID: 4HR9) were preprocessed using Discovery Studio and visualized in PyMOL (Additional file 1: Fig. S6B and S6C). The interactions are illustrated in Additional file 1: Fig. S6D, where bsscFv is shown in green, BAFF in blue, and IL-17 in purple. Yellow dashed lines indicate hydrogen bonds, while red dashed lines represent hydrophobic interactions. The binding scores for bsscFv-BAFF and bsscFv-IL-17 are −295.89 and −288.00, respectively, with confidence scores of 94%, indicating strong binding stability. Key interactions include hydrogen bonds between BAFF’s MET236/THR239 and bsscFv’s TYR176/TYR103 (3.1 Å, 3.0 Å). For IL-17, ASN25 forms a hydrogen bond with bsscFv’s THR320 (2.7 Å), and ARG20 of IL-17 forms three hydrogen bonds with bsscFv’s TYR363, ASN365, and TYR367 (2.3 Å, 2.5 Å, 3.1 Å). In Additional file 1: Fig. S6E, "A" represents bsscFv and "B" represents BAFF, while Additional file 1: Fig. S6F shows "A" as bsscFv and "B" as IL-17, further confirming these binding interactions. These interactions are critical for the stable binding between the bsscFv, BAFF, and IL-17 proteins.

Survival analysis of mice during the experimental period

Kaplan–Meier survival analysis was conducted to evaluate the mortality of mice across all in vivo experimental groups, as shown in Supplementary Additional file 1: Fig. S7. Throughout the treatment period, all groups were monitored for survival, and no deaths were observed in any cohort that could be attributed to treatment-related toxicity or disease progression. Statistical analysis using the log-rank (Mantel–Cox) test revealed no significant differences in survival rates among the groups (P > 0.05). These findings indicate that administration of the bispecific antibody, across the tested doses and experimental conditions, did not adversely affect survival and was well tolerated in vivo.

Discussion

This study presents the development and evaluation of a bsscFv targeting BAFF and IL-17 for the treatment of SLE. The bsscFv was engineered by fusing scFvs specific to BAFF and IL-17 via a flexible linker, yielding low-nanomolar binding affinities for both targets, comparable to their parental antibodies.

In vitro assays demonstrated potent dual neutralization by the bsscFv, effectively inhibiting BAFF- and IL-17-mediated B-cell survival, proliferation, immunoglobulin production, and proinflammatory cytokine secretion by peripheral blood mononuclear cells. These findings underscore the dual capacity of bsscFv to disrupt two critical immunopathological axes in SLE: autoreactive B-cell activation and systemic inflammation.

In vivo, bsscFv treatment in the MRL/lpr murine model—recapitulating major features of human SLE—led to dose-dependent reductions in autoantibody titers, proteinuria, and glomerulonephritis. These effects were accompanied by a broad suppression of proinflammatory cytokines (IL-2, IL-4, IL-6, IL-10, IFN-α, and TNF-α) in both kidney and spleen tissues, reinforcing the therapeutic potential of dual-pathway blockade.

When compared to current therapeutic strategies, the bsscFv offers several potential advantages. For instance, belimumab, a monoclonal antibody targeting BAFF, has been approved for SLE but exhibits limited efficacy in patients with high disease activity or refractory manifestations. Similarly, IL-17-targeted therapies such as secukinumab have shown promise in autoimmune diseases like psoriasis and ankylosing spondylitis, but their role in SLE remains exploratory and is often limited by partial responses. Moreover, dual therapy combining BAFF and IL-17 inhibitors may enhance disease control but is associated with increased cost, complexity, potential for overlapping toxicities, and reduced patient adherence.

In contrast, the bsscFv format provides a streamlined and mechanistically synergistic approach, integrating dual specificity into a single molecular entity. This may result in more efficient immune modulation, favorable pharmacokinetics, reduced dosing frequency, and better patient compliance [23]. Furthermore, bispecific antibodies have been shown to induce cooperative effects that exceed those of monotherapies or their combinations, possibly by simultaneously engaging spatially or temporally linked immune mechanisms.

The favorable safety signals observed in preliminary non-human primate studies further support the clinical translatability of the bsscFv. Nonetheless, this study has limitations. The therapeutic efficacy was assessed in a single murine model, and the cellular selectivity of bsscFv remains incompletely characterized. While cytokine inhibition and disease amelioration were clearly demonstrated, the preferential targeting of autoreactive versus non-pathogenic lymphocyte subsets was not directly assessed.

Encouragingly, ongoing work (manuscript in preparation) suggests that bsscFv treatment selectively reduces pathogenic cytokine production and autoantibody levels, with minimal impact on total immunoglobulin levels and naïve lymphocyte populations. These data imply a degree of immunological precision, potentially arising from elevated expression of BAFF and IL-17 in activated immune niches.

To further address these critical questions, future studies should incorporate high-resolution profiling approaches, such as single-cell RNA sequencing or multi-parametric flow cytometry, to delineate bsscFv’s cellular targets in murine and human immune systems. Additionally, comprehensive preclinical studies evaluating long-term safety, efficacy, and immunogenicity are necessary before clinical translation.

In conclusion, our findings provide compelling evidence that bispecific targeting of BAFF and IL-17 offers a promising therapeutic approach for SLE by simultaneously modulating B-cell-driven autoimmunity and inflammatory cytokine networks. Compared to existing monotherapies and combination regimens, the bsscFv holds distinct advantages in molecular efficiency, breadth of action, and potential clinical convenience. Further mechanistic and translational investigations are warranted to establish its safety, specificity, and therapeutic durability in human SLE.

Conclusions

In this study, we successfully engineered and characterized a bsscFv antibody targeting BAFF and IL-17, two pivotal cytokines implicated in the pathogenesis of SLE. The bsscFv exhibited robust binding affinity to both targets with minimal cross-interference, and effectively neutralized BAFF- and IL-17-mediated biological activities in vitro. In vivo administration of bsscFv in MRL/lpr mice led to significant reductions in autoantibody levels, proteinuria, renal pathology, and inflammatory cytokine expression, accompanied by histopathological improvements and decreased immune complex deposition in kidney tissues. PK and PD analyses confirmed its favorable bioavailability, biological activity, and lack of observable toxicity. Multi-omics analyses further supported the therapeutic impact of bsscFv on immune regulation and metabolic rebalancing. Taken together, these results indicate that bsscFv is a promising bispecific therapeutic candidate for the treatment of SLE, meriting further preclinical and clinical development.

Methods

Vector construction and cell culture

The coding sequences of human BAFF (UniProt: Q9Y275, residues A134–L285) and IL-17A (UniProt: Q16552, residues G24–A155) were amplified by PCR and cloned into the pCDNA3.1(+) vector (Thermo Fisher). The plasmids were transfected into HEK 293 F cells using polyethylenimine (PEI, Polysciences, 1 mg/mL, pH 7.0) at a DNA:PEI ratio of 1:3. Transfected cells were cultured in FreeStyle™ 293 Expression Medium (Gibco) for 72 h at 37 °C, 5% CO₂ with shaking (120 rpm). Supernatants were harvested by centrifugation at 4000 × g for 15 min. The scFv variable regions targeting BAFF and IL-17 were cloned into pET-28a(+) (Novagen), with a flexible (G₄S)₂ linker, to construct bispecific scFv (bsscFv). Plasmids were transformed into E. coli BL21(DE3) (TransGen Biotech), cultured in LB medium with 50 μg/mL kanamycin, induced with 0.5 mM IPTG at 16 °C for 16 h when OD600 reached 0.6. Raji and HT-29 cells were cultured in RPMI-1640 and McCoy’s 5 A media (Gibco), respectively, supplemented with 10% fetal bovine serum (FBS, Gibco) and 1% penicillin–streptomycin (100 U/mL penicillin, 100 μg/mL streptomycin) at 37 °C in a 5% CO₂ incubator.

Protein purification and expression

Bacterial pellets were resuspended in lysis buffer (50 mM Tris–HCl, pH 8.0; 300 mM NaCl; 10 mM imidazole; 1 mM PMSF), sonicated on ice (10 s on/20 s off cycles for 10 min), and centrifuged at 12,000 × g for 30 min at 4°C. The supernatant was applied to a Ni–NTA agarose column (Qiagen) pre-equilibrated with the same buffer. After washing with wash buffer (50 mM Tris–HCl, 300 mM NaCl, 30 mM imidazole, pH 8.0), bound proteins were eluted with elution buffer (50 mM Tris–HCl, 300 mM NaCl, 250 mM imidazole, pH 8.0). Proteins were dialyzed against PBS (pH 7.4) and concentrated using Amicon Ultra-15 centrifugal filters (30 kDa cutoff, Millipore). Protein concentration was quantified by Bradford assay (Bio-Rad), and purity was confirmed via 12% SDS-PAGE.

Western blot

Proteins (10 μg/lane) were separated by SDS-PAGE and transferred to a 0.45 μm PVDF membrane (Millipore) using a semi-dry blotting system (Bio-Rad). Membranes were blocked with 5% non-fat milk in TBST (Tris-buffered saline with 0.1% Tween-20) for 1 h at room temperature. Primary antibody: anti-His monoclonal antibody (1:5000, Abcam, ab18184), incubated overnight at 4°C. After washing, membranes were incubated with HRP-conjugated goat anti-mouse IgG (1:5000, Abcam, ab6789) for 1 h at room temperature. Signal was developed using the ECL Plus detection kit (Tanon).

Mouse immunization protocol and spleen cell isolation

Female BALB/c mice (6–8 weeks old) were purchased from the Zhengzhou University Experimental Animal Center and the Henan Province Experimental Animal Center. All animal experiments were conducted in accordance with the guidelines of the Animal Ethics Committee of Zhengzhou University, and the study protocol was approved by the committee (Approval No. ZZUIRB2021-141). Prior to immunization, mice were anesthetized using isoflurane inhalation (4% for induction, 2% for maintenance) to minimize discomfort. At the study endpoint, euthanasia was performed by CO₂ inhalation, in line with the AVMA Guidelines for the Euthanasia of Animals (2020). The animals were immunized with recombinant BAFF (50 μg) and IL-17 (50 μg) proteins in CFA on day 0, followed by booster immunizations on days 14 and 28 using IFA. On day 35, blood samples were collected for antibody titers. A final booster (50 μg BAFF and IL-17) was administered on day 42. Three days later, mice were euthanized, and spleens were harvested, disrupted, filtered, and centrifuged. Spleen cell suspensions, rather than purified B lymphocytes, were used in downstream assays to preserve the native immunological microenvironment and maintain physiological cell–cell interactions essential for cytokine-driven immune responses, including T cell help, antigen presentation, and cytokine crosstalk. This approach provides a more biologically relevant model for evaluating the activation and transcriptional responses of B cells within the splenic milieu. Cells were treated with RBC lysis buffer, resuspended in PBS, counted using trypan blue, and prepared for RNA extraction and cDNA synthesis (Fig. 6A).

Fig. 6
figure 6

Schematic representation of mouse immunization and single-chain antibody screening. A Mouse immunization protocol. B Schematic of phage display selection. C Analysis of specific antibody sequences

mRNA extraction, reverse transcription, and qPCR

Total RNA was extracted using the FastPure® Cell/Tissue Total RNA Isolation Kit (Vazyme RC101), quantified with a NanoDrop 2000 spectrophotometer (Thermo). Reverse transcription was performed using the PrimeScript™ RT reagent Kit (Takara). qPCR was conducted using ChamQ SYBR qPCR Master Mix (Vazyme Q331) on a LightCycler 96 system (Roche). Gene expression levels were normalized to GAPDH, and relative changes calculated using the 2−(ΔΔCt) method [24].

PCR amplification of antibody light and heavy chain variable regions

The antibody light (VL) and heavy (VH) chain variable regions were amplified from cDNA using PCR with degenerate primers (Additional file 1: Table 1) based on mouse immunoglobulin sequences. The PCR mixture (50 μL) included 1 × buffer, 2.5 mM MgCl2, 0.2 mM dNTPs, 0.5 μM primers, 1 U Taq polymerase (Takara), and 2 μL cDNA. PCR conditions: 95 °C for 5 min, 30 cycles (95°C for 30 s, 58 °C for 30 s, 72 °C for 1 min), final extension at 72 °C for 5 min. PCR products were purified (Tiangen kit) and analyzed by agarose gel electrophoresis.

Analysis of scFv insertion rate and antibody library sequence diversity

Insertion Rate: Forty-eight DH5α/pCANTAB 5E single colonies were selected for PCR amplification to evaluate the insertion rate of scFvs based on the presence or absence of PCR products. Antibody Sequence Diversity: Next-generation sequencing (NGS) was used to assess the diversity of the antibody library sequences.

M13K07 helper phage amplification and titer determination

M13K07 phage (Fenghui Biotech) was amplified in E. coli TG1 cells (Beijing Huayueyang Biotechnology) in 2 × YT medium with 50 μg/mL kanamycin. The culture was incubated at 37 °C until OD600 reached 0.5, then phage was added at MOI 20 and incubated for 1 h. Infected cells were grown overnight at 30°C. Phage was precipitated using 20% PEG/2.5 M NaCl, resuspended in PBS, and titered by infecting E. coli TG1 cells with serial dilutions and plating on LB agar with kanamycin.

Construction and quality assessment of scFv library

The scFv library was constructed by linking VL and VH sequences with a (G4S)4 linker via overlap extension PCR. The fragments were digested with BamHI and XhoI, ligated into pCANTAB 5E, and transformed into E. coli TG1 cells. Library size was estimated by colony count. Random clones were screened by PCR with pCANTAB 5E-specific primers and analyzed by gel electrophoresis to confirm full-length scFv inserts.

Phage display selection

The scFv library was biopanned against recombinant human BAFF and IL-17 proteins over five rounds. In each round, plates were coated with 100 μL of antigen (10 μg/mL) overnight at 4 °C, blocked with 3% BSA, and incubated with phage library for 2 h at 37°C. Bound phages were eluted with 100 μL of 100 mM glycine–HCl (pH 2.2), neutralized, and used to infect TG1 cells. After five rounds, individual clones were selected for phage ELISA to identify antigen-specific binders (Fig. 6B and C).

Enzyme-linked immunosorbent assay (ELISA)

Phage ELISA was used to assess scFv binding. Plates were coated with recombinant BAFF or IL-17 (2 μg/mL) overnight at 4°C. After blocking with 3% BSA, phage supernatants were added for 1 h. After washing, HRP-conjugated anti-M13 antibody was added, developed with TMB, and stopped with 2 M H2SO4. Absorbance at 450 nm was measured, and clones binding to both BAFF and IL-17 were selected for further study.

Construction and expression of bsscFv

Anti-BAFF and anti-IL-17 scFv sequences were linked by bridging PCR to form bsscFv. Overlapping primers amplified each scFv, and fragments were combined in a second PCR to produce full-length bsscFv. The construct was cloned into the pET-28a(+) vector, transformed into BL21(DE3) cells, and expressed. Protein was purified using Ni–NTA affinity chromatography, as described in the "Protein Purification and Expression" section.

Affinity determination

The binding kinetics of the bsscFv antibody to human BAFF and IL-17 were evaluated using a label-free biomolecular interaction analysis system (Gator Prime, Gator Bio, Suzhou, China), following the manufacturer’s instructions. Biotinylated scFv and bsscFv antibodies were individually immobilized onto streptavidin-coated biosensors. Serial dilutions of recombinant human BAFF or IL-17 were introduced as analytes to determine the association rate constant (ka) and dissociation rate constant (kd), from which the equilibrium dissociation constant (KD) was derived. To assess potential binding interference between the two targets, additional kinetic analyses were conducted under antigen-saturation conditions. Specifically, bsscFv-coated biosensors were pre-incubated with an excess of BAFF prior to IL-17 binding measurements, and vice versa. All experiments were performed in triplicate, and kinetic parameters were analyzed using Gator Analysis software based on a global 1:1 binding model [25].

In vitro experiments

The bioactivities of recombinant BAFF and IL-17 were evaluated by assessing their effects on Raji cell proliferation and CXCL1 secretion in HT-29 cells [26, 27]. Cells (1 × 106 cells/mL) were stimulated with various concentrations of BAFF or IL-17 (0.1 to 100 nM) for 72 h. Raji cell counts and CXCL1 levels were measured. For inhibition assays, the effects of BAFF and IL-17 were tested in the presence of each other with an irrelevant scFv as a negative control. The percentage of inhibition was calculated based on the minimal (no protein) and maximal (150 pM BAFF or 4 nM IL-17) responses. IC50 values were determined using GraphPad Prism.

In vivo experiments

Female BALB/c mice (10–12 weeks old, n = 5 per group) were randomly assigned to eight groups to evaluate the in vivo neutralizing activity of scFvs and the bispecific bsscFv against BAFF and IL-17, respectively. For BAFF-related experiments, four groups were included: one group received anti-BAFF scFv1 (5 nM/mouse), one received bispecific bsscFv (5 nM/mouse), and two groups received no antibody. After 48 h, one of the antibody-free groups remained untreated as a baseline control (NC), while the other was administered recombinant human BAFF (1 nM/mouse) via intraperitoneal injection. Mice were euthanized six days after BAFF administration, and spleens were collected for quantification of CD19 mRNA expression by qPCR as a marker of B cell activation. Similarly, four groups were used for IL-17-related experiments: one group received anti-IL-17 scFv2 (5 nM/mouse), one received bsscFv (5 nM/mouse), and two received no antibody. After 48 h, one control group remained untreated (NC), and the other was administered recombinant human IL-17 (0.5 nM/mouse) intraperitoneally. Mice were sacrificed two hours later, and serum samples were collected to measure CXCL1 protein levels by ELISA as a downstream indicator of IL-17 signaling. This experimental setup enabled a direct comparison of the monospecific and bispecific scFvs in suppressing cytokine-mediated immune responses in vivo [28].

Pharmacokinetics/pharmacodynamics (PK/PD)

Female BALB/c mice were randomly divided into four groups (n = 5 per group) and administered intravenous injections of bsscFv at doses of 1, 5, or 10 mg/kg, or PBS as a control. PK samples were collected at 0.25, 8, and 24 h post-injection, and subsequently once per week for up to 91 days. Serum was used to coat BAFF and IL-17 antigens onto 96-well plates, detected with HRP-labeled anti-G4S antibody. OD450nm was measured to determine bsscFv concentration by BCA protein assay. PK data were analyzed for Cmax, AUC0-∞, clearance (CL), and half-life (T1/2). BAFF and IL-17 levels were measured by ELISA. For PD, mRNA was extracted from serum collected pre-dose (days −6, −1) and on subsequent study days. CD19 expression was quantified by qPCR and changes were calculated from baseline levels.

Mouse disease model (MRL/lpr) experiments

Female MRL/lpr mice (10–12 weeks old, n = 5 per group) were used to evaluate the therapeutic efficacy of the bsscFv antibody. Three experimental groups received bsscFv via intravenous injection at doses of 1 mg/kg, 5 mg/kg, or 10 mg/kg, representing low, medium, and high treatment groups, respectively. Two control groups were included: one consisting of age-matched BALB/c mice (healthy control, 10–12 weeks old, 5 per group), and the other of untreated MRL/lpr mice (disease control, 10–12 weeks old, 5 per group). At the study endpoint, blood samples were collected, and serum levels of blood urea nitrogen (BUN), double-stranded DNA antibodies (dsDNA Ab), serum creatinine (Scr), antinuclear antibodies (ANA), small nuclear ribonucleoprotein/Smith antigen (snRNP/SM), and systemic lupus erythematosus-related IgG (SLE-IgG) were measured by ELISA, following the manufacturer's protocols. Kidney tissues were collected and processed for multiple analyses. Total RNA was extracted from renal tissue using the Vazyme FastPure® RNA kit, and mRNA expression levels of TNF-α, IFN-α, IL-2, IL-4, IL-6, and IL-10 were quantified by qPCR using GAPDH as the internal control. Protein expression levels of these cytokines were further assessed by Western blotting, as described in Section "PCR of antibody chains and construction of scFv phage library".

For histopathological evaluation, kidney sections were fixed in 4% paraformaldehyde, embedded in paraffin, and stained with hematoxylin and eosin (H&E) for the assessment of glomerular and tubular morphology, including sclerosis and inflammatory infiltration. Additional sections were stained with Periodic Acid–Schiff (PAS) and Masson’s Trichrome to evaluate polysaccharide accumulation and collagen fiber deposition, respectively. Quantitative analysis was performed using Image-Pro Plus 6.0 software. To assess renal immune complex deposition and histopathological alterations, frozen kidney sections were stained with FITC-conjugated goat anti-mouse IgG (H + L) antibody (Jackson ImmunoResearch, USA) following standard immunofluorescence procedures. Sections were imaged using a fluorescence microscope (Nikon Eclipse C1), and the fluorescence intensity and IgG-positive area were quantified using ImageJ software. Results were compared across experimental and control groups.

Transcriptomic and metabolomic analysis of kidneys

Kidney tissues from three MRL/lpr mice per group were collected for analysis. Total RNA was extracted for transcriptomic analysis via RNA sequencing (RNA-seq) to identify differentially expressed genes. For metabolomic analysis, kidney samples were processed and analyzed by LC–MS to profile metabolic changes and identify differential metabolites. The results from both analyses were integrated to assess the impact of treatment on gene expression and metabolic pathways.

Analysis of binding sites

Protein–protein docking was performed using ZDOCK in Discovery Studio. The bsscFv structure was predicted by AlphaFold, while BAFF (1KD7) and IL-17 (4HR9) structures were sourced from the Protein Data Bank. bsscFv served as the receptor, with BAFF and IL-17 as ligands. An angular step size of 6 generated 54,000 conformations, which were filtered by binding site and RMSD relative to POSE36. Binding sites with RMSD < 15 were refined using RDOCK for energy minimization and scoring based on the CHARMm energy function.

Data analysis

Data are presented as mean ± SEM. Statistical significance was assessed using one-way ANOVA with Tukey's post hoc test for multiple comparisons or Student's t-test for pairwise comparisons. Survival curves were analyzed by the Kaplan–Meier method and compared using the log-rank test. Pearson's correlation coefficient was used to assess correlations. *P < 0.05, P < 0.01, *P < 0.001; ns, not significant. All analyses were performed using GraphPad Prism software (version 8.0).

Data availability

Data will be made available on request.

Abbreviations

ANA:

Antinuclear antibodies

BAFF:

B-cell activating factor

BUN:

Blood urea nitrogen

bsscFv:

Bispecific single-chain variable fragment

dsDNA Ab:

Double-stranded DNA antibodies

ELISA:

Enzyme-linked immunosorbent assay

H&E:

Hematoxylin and eosin

IL-17:

Interleukin-17

M:

MRL/lpr model

NGS:

Next-generation sequencing

PAS:

Periodic Acid–Schiff

PD:

Pharmacodynamic

PK:

Pharmacokinetic

RNA-seq:

RNA sequencing

SLE:

Systemic lupus erythematosus

SLE-IgG:

Systemic lupus erythematosus-related IgG

scFvs:

Single-chain variable fragments

Scr:

Serum creatinine

snRNP/Sm:

Small nuclear ribonucleoprotein/Smith antigen

T:

Bispecific antibody-treated group

Th17:

T helper 17

TNF:

Tumor necrosis factor

References

  1. Wang FQ, Dang X, Yang WL. Transcriptomic studies unravel the molecular and cellular complexity of systemic lupus erythematosus: A review. Clin Immunol. 2024;268:110367.

  2. Kaul A, Gordon C, Crow MK, Touma Z, Urowitz MB, van Vollenhoven R, et al. Systemic lupus erythematosus. Nat Rev Dis Primers. 2016;2:16039.

  3. Ameer MA, Chaudhry H, Mushtaq J, Khan OS, Babar M, Hashim T, et al. An Overview of Systemic Lupus Erythematosus (SLE) Pathogenesis, Classification, and Management. Cureus J Med Science. 2022;14(10):e30330.

  4. Goulielmos GN, Zervou MI, Vazgiourakis VM, Ghodke-Puranik Y, Garyfallos A, Niewold TB. The genetics and molecular pathogenesis of systemic lupus erythematosus (SLE) in populations of different ancestry. Gene. 2018;668:59–72.

    Article  PubMed  Google Scholar 

  5. Luo SHT, Long H, Lu QJ. Recent advances in understanding pathogenesis and therapeutic strategies of systemic lupus erythematosus. Int Immunopharmacol. 2020. https://doi.org/10.1016/j.intimp.2020.107028.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Benschop RJ, Chow CK, Tian Y, Nelson J, Barmettler B, Atwell S, et al. Development of tibulizumab, a tetravalent bispecific antibody targeting BAFF and IL-17A for the treatment of autoimmune disease. MAbs. 2019;11(6):1175–90.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Möckel T, Basta F, Weinmann-Menke J, Schwarting A. B cell activating factor (BAFF): structure, functions, autoimmunity and clinical implications in systemic lupus erythematosus (SLE). Autoimmun Rev. 2021. https://doi.org/10.1016/j.autrev.2020.102736.

    Article  PubMed  Google Scholar 

  8. Martin JC, Baeten DL, Josien R. Emerging role of IL-17 and Th17 cells in systemic lupus erythematosus. Clin Immunol. 2014;154(1):1–12.

    Article  PubMed  Google Scholar 

  9. Schneider P, MacKay F, Steiner V, Hofmann K, Bodmer JL, Holler N, et al. BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth. J Exp Med. 1999;189(11):1747–56.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mackay F, Browning JL. BAFF: a fundamental survival factor for B cells. Nat Rev Immunol. 2002;2(7):465–75.

    Article  PubMed  Google Scholar 

  11. Samy E, Wax S, Huard B, Hess H, Schneider P. Targeting BAFF and APRIL in systemic lupus erythematosus and other antibody-associated diseases. Int Rev Immunol. 2017;36(1):3–19.

    Article  PubMed  Google Scholar 

  12. Zenobia C, Hajishengallis G. Basic biology and role of interleukin-17 in immunity and inflammation. Periodontol 2000. 2015;69(1):142–59.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Xu S, Cao XT. Interleukin-17 and its expanding biological functions. Cell Mol Immunol. 2010;7(3):164–74.

    Article  PubMed  PubMed Central  Google Scholar 

  14. López P, Rodríguez-Carrio J, Caminal-Montero L, Mozo L, Suárez A. A pathogenic IFNα, BLyS and IL-17 axis in systemic lupus erythematosus patients. Sci Rep. 2016. https://doi.org/10.1038/srep20651.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Doreau A, Belot A, Bastid J, Riche B, Trescol-Biemont MC, Ranchin B, et al. Interleukin 17 acts in synergy with B cell-activating factor to influence B cell biology and the pathophysiology of systemic lupus erythematosus (vol 10, pg 778, 2009). Nat Immunol. 2014;15(9):894.

  16. Thorley J. Belimumab and rituximab for SLE. Lancet Rheumatol. 2024;6(10):e666-e.

    Article  Google Scholar 

  17. Abid N, Manaye S, Naushad H, Cheran K, Murthy C, Bornemann EA, et al. The Safety and Efficacy of Rituximab and Belimumab in Systemic Lupus Erythematosus: A Systematic Review. Cureus J Med Science. 2023;15(6):e40719.

  18. Nalbandian A, Crispín JC, Tsokos GC. Interleukin-17 and systemic lupus erythematosus: current concepts. Clin Exp Immunol. 2009;157(2):209–15.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Crispín JC, Kyttaris VC, Tsokos GC. The Role of Interleukin-17 in Systemic Lupus Erythematosus. Th 17 Cells Health Dis. 2011:391–400.

  20. Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs. 2017;9(2):182–212.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Fan GW, Wang ZJ, Hao MJ, Li JM. Bispecific antibodies and their applications. J Hematol Oncol. 2015;8:130.

  22. Thakur A, Huang M, Lum LG. Bispecific antibody based therapeutics: strengths and challenges. Blood Rev. 2018;32(4):339–47.

    Article  PubMed  Google Scholar 

  23. Spiess C, Zhai QT, Carter PJ. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol. 2015;67(2):95–106.

    Article  PubMed  Google Scholar 

  24. Karlen Y, McNair A, Perseguers S, Mazza C, Mermod N. Statistical significance of quantitative PCR. BMC Bioinformatics. 2007. https://doi.org/10.1186/1471-2105-8-131.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Raghava GPS, Agrewala JN. Method for determining the affinity of monoclonal-antibody using noncompetitive Elisa - a computer-program. J Immunoassay. 1994;15(2):115–28.

    Article  PubMed  Google Scholar 

  26. Liang DF, Zeng QY, Xu ZG, Zhang H, Gui L, Xu C, et al. BAFF activates Erk1/2 promoting cell proliferation and survival by Ca-CaMKII-dependent inhibition of PP2A in normal and neoplastic B-lymphoid cells. Biochem Pharmacol. 2014;87(2):332–43.

    Article  PubMed  Google Scholar 

  27. Straus DS. TNFα and IL-17 cooperatively stimulate glucose metabolism and growth factor production in human colorectal cancer cells. Mol Cancer. 2013. https://doi.org/10.1186/1476-4598-12-78.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Depoil D, Fleire S, Treanor BL, Weber M, Harwood NE, Marchbank KL, et al. CD19 is essential for B cell activation by promoting B cell receptor-antigen microcluster formation in response to membrane-bound ligand. Nat Immunol. 2008;9(1):63–72.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the members of the Molecular Immunology Laboratory at Zhengzhou University, for their assistance with the experiments.

Funding

This study was supported by research funds from the Major Science and Technology Projects in Henan Province, Innovation Research on Antibody and Protein-based Drugs (Project No. 241110310100).

Author information

Authors and Affiliations

Authors

Contributions

CX: Writing Original draft preparation, Data curation, Visualization, Investigation. JMZ, YMC, YKL, HLL, CL, XFZ: Methodology, Supervision. YHQ: Supervision. GPZ and APW: Conceptualization, Methodology, Data curation, Software, Reviewing. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Gaiping Zhang or Aiping Wang.

Ethics declarations

Ethics approval and consent to participate

All animal experiments were conducted in accordance with the guidelines of the Animal Ethics Committee of Zhengzhou University, and the study protocol was approved by the committee (Approval No. ZZUIRB2021-141).

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

12915_2025_2398_MOESM1_ESM.pdf

Additional file 1: Figures S1-S7; Table 1-5. Fig. S1. Protein purification of BAFF and IL-17 and immunogenicity evaluation in mice. Fig. S2. PCR amplification of antibody variable regions and recombinant scFv construction. Fig. S3. Sequence diversity analysis of the antibody library. Fig. S4. Phage library panning results. Fig. S5. Construction and affinity assessment of bispecific single-chain antibodies. Fig. S6. Analysis of antigen-antibody binding sites. Fig. S7. Kaplan-Meier survival curves of mice from in vivo experiments. Table 1. Primer Sequences. Table 2. Count per plate of scFv library. Table 3. Counts per plate for helper phage M13K07. Table 4. ScFv library capacity determination. Table 5. Selective enrichment of scFv during screening.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xin, C., Zhou, J., Chen, Y. et al. Design and development of a bispecific antibody targeting BAFF and IL-17 for systemic lupus erythematosus treatment. BMC Biol 23, 283 (2025). https://doi.org/10.1186/s12915-025-02398-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12915-025-02398-2

Keywords