Next Article in Journal
Curcumin Microcapsule Formulations for Prolong Persistence in the Photodynamic Inactivation of Aedes aegypti Larvae
Previous Article in Journal
Multi-Omics Analysis of the Immune Effect of the Engineered Exosome Drug Delivery System in Inducing Macrophage Apoptosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecule Formats of B-Cell Targeting Biologics: Applications in Autoimmune Disease Treatment and Impacts on Manufacturability

by
Yueming Qian
1,* and
Tamer I. Mahmoud
2
1
Pre-Pivotal Drug Substance Technologies, Amgen Inc., Rockville, MD 20850, USA
2
Discovery Research, Amgen Inc., Rockville, MD 20850, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(4), 495; https://doi.org/10.3390/pharmaceutics17040495
Submission received: 17 March 2025 / Accepted: 4 April 2025 / Published: 8 April 2025
(This article belongs to the Section Biopharmaceutics)

Abstract

:
The targeting of B-lymphocyte cells has emerged as one of the most pivotal strategies in the management of autoimmune diseases. This review provides an overview of protein therapeutics illustrating their direct and indirect effects on B-cells using different molecule formats. The design and format of these molecules influence their mode of action and affect their manufacturing strategies. Manufacturability should be assessed at an early stage and continuously through collaboration between discovery and development teams. Scalability evaluations should encompass not only process development and facility compatibility but also cell line development. Examples of format-specific manufacturability of biologics are reviewed to offer general insights into enhancing productivity and quality in a cost-effective manner.

1. Introduction

The immune system includes the innate and adaptive subsystems, which work together to defend against infection and disease. In autoimmune diseases, the immune system may malfunction, erroneously targeting its own cells, tissues, and organs. The NIH Autoimmune Disease Coordinating Committee reports that there are at least 80 types of chronic and often disabling autoimmune disorders, affecting about 24 million people in the United States (https://dpcpsi.nih.gov/sites/default/files/NIH-Triennial-Report-FY-2016-2018_Final508.pdf, accessed on 3 April 2025). The root cause remains poorly understood, but an overactive response is frequently observed in the immune network, involving humoral, cellular components, or both.
B-cells are vital for humoral immunity, but they can become pathogenic in autoimmunity [1]. Autoreactive B-cells may produce autoantibodies against the body’s tissues, present self-antigens to T-cells, or generate inflammatory cytokines. Due to their role in autoimmune disease initiation and progression [2], B-cells have become key therapeutic targets. Strategies to target B-cells vary based on the disease mechanism and B-cell functions [3]. B-cell depletion effectively treats pemphigus and neuromyelitis optica spectrum disorder (NMOSD) [4] but not for other autoimmune diseases, such as systemic lupus erythematosus (SLE) [3]. The variability in efficacy may be associated with the breadth and depth of pathogenic B-cell depletion and the diversity of B-cell functions being a driver of pathophysiology in these heterogeneous diseases.
There are different methods to target B-cells [5], such as cell-based and biologics-based therapeutics. Targeting B-cells by biologics can be direct or indirect. Direct depletion is achieved by targeting surface molecules like CD20, CD19, and B-cell-activating factor receptors (BAFF-R) using monoclonal antibodies. Indirect depletion involves blocking survival cytokines or regulators of activation and differentiation. BAFF/BAFF-R interaction promotes survival gene expressions in B-cells and plasma cells where BAFF-R is a direct target and BAFF is an indirect target. CD40 and CD40L are another pair of direct and indirect B-cell targets; blocking their interaction inhibits B-cell gene transcription, cell growth, and differentiation into memory B cells and plasma cells. Other modalities, such as Chimeric Antigen Receptor (CAR) T-cell therapy, have also been explored to target B-cells; however, the following sections will only discuss protein therapeutics that target/interact with B-cells directly or indirectly. The focus will be on the impacts of biologics formats on their mode of action and manufacturability. This topic, yet to be reviewed, is crucial for advancing biologics development from discovery through to manufacturing.

2. Different Molecule Formats and Features Applied in Biologics Development for Autoimmune Disease Treatments

Previous review articles have collectively provided a good reference for B-cell targeting biologics from different perspectives [6,7,8,9]. Table 1 summarizes the related contents with additional molecules, new updates, and the highlights of molecule formats and features. Six representative products are discussed relating to different molecule formats and features applied in biologics development and their mode of action in the associated diseases.

2.1. Ocrelizumab and Ofatumumab—Next Generation Anti-CD20 Monoclonal Antibodies (mAbs)

Both ocrelizumab and ofatumumab, as examples of next-generation B-cell targeting mAbs, were designed to reduce immunogenicity with the former humanized and the latter fully human. Ocrelizumab binds an epitope that overlaps with rituximab’s binding site and offers enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) [7] whereas ofatumumab specifically recognizes an epitope that encompasses both the small and large extracellular loops of CD20 and has a more effective complement-dependent cytotoxicity (CDC) induction and killing target cell capacity [32]. Ocrelizumab was first approved in the US for the treatment of patients with relapsing–remitting multiple sclerosis (RRMS) and primary progressive multiple sclerosis (PPMS) in 2017 [33], which led to B cell depletion being a mainstay of treatment for MS, and the first therapy specifically for PPMS [34]. In addition to the approvals for RRMS, PPMS, and secondary progressive MS (SPMS) [35], Ofatumumab is also under clinical investigation for RA.

2.2. Inebilizumab—Afucosylated Anti-CD19 mAb

Developed by Viela Bio and acquired by Horizon Therapeutics and more recently by Amgen, inebilizumab is a humanized IgG1 mAb targeting the extracellular loop of CD19 [36,37], It initially received FDA approval in 2020 for the treatment of NMOSD in adult patients who are seropositive for immunoglobulin G (IgG) autoantibodies against aquaporin-4 (AQP4) [4]. Since then, the authorization for access to this biologic drug has been quickly expanded to include NMOSD patients in Japan in 2021, in Germany and France in August 2022, and in Brazil in December 2022 [38]. As an afucosylated IgG1, inebilizumab has a 10-fold higher binding affinity to human Fcγ receptor (FcγR) IIIA and achieves quick and potent B-cell depletion through ADCC [36]. Further clinical data showed that the beneficial outcomes could be maintained for more than 4 years in terms of the reductions in NMOSD attacks [39] and in AQP4-IgG titers. Other indications such as kidney transplant desensitization, myasthenia gravis, and IgG4-RD are also under clinical evaluation [4]. Phase 3 clinical trial for the treatment of IgG4-RD was a randomized, double-blind, and placebo-controlled study at 80 sites in 22 countries. Compared to the placebo for its primary endpoint, this novel and the steroid-sparing trial showed a statistically significant 87% reduction in the risk of IgG4-RD flare for 52 weeks; all key secondary endpoints including annualized flare rate and treatment-free and corticosteroid-free complete remission were all met [14]. In a Phase 3 trial in myasthenia gravis (NCT04524273), inebilizumab met its primary endpoint, with a statistically significant change from baseline in Myasthenia Gravis Activities of Daily Living (MG-ADL) score (−4.2) compared with placebo (−2.2) (difference: −1.9, p < 0.0001) at Week 26 for the combined study population, and demonstrated continued improvement through this period of time (https://wwwext.amgen.com/newsroom/press-releases/2024/10/amgen-presents-positive-phase-3-data-for-uplizna-inebilizumabcdon-in-generalized-myasthenia-gravis-gmg-at-aanem-2024, accessed on 15 March 2025).

2.3. Belimumab—mAb Indirectly Targeting B Cells

Belimumab is a fully human mAb that does not directly target B-cells but neutralizes the biologically active soluble form of BAFF and in turn, blocks the binding of this cytokine stimulator to its receptor on the involved B-cells [20]. It has been found that BAFF overexpression induces autoreactive B cells to increase autoantibody levels under autoimmune conditions [40]. Inhibition of B-cell proliferation and differentiation by belimumab represents a treatment option for adult patients with seropositive active SLE (especially musculoskeletal and cutaneous disease). Following the FDA approval of belimumab in 2011, which was the first biologic approved for SLE and the first new lupus drug in more than 50 years [20], BAFF was established as a molecular target for further therapeutic developments. Telitacicept is an Fc fusion protein that contains TACI amino acids 13-118 of the extracellular domain, which binds to and neutralizes BAFF and APRIL. It received approval for SLE in China in 2021 [29]. Conversely, tabalumab is a humanized monoclonal antibody designed to have a broader blocking effect by neutralizing both soluble and membrane-bound BAFF. However, it did not achieve the expected clinical results in relapsing multiple sclerosis [41,42]. Furthermore, atacicept is another TACI-Ig fusion protein that binds to and blocks BAFF and APRIL [41]. Blisibimod, an Fc-conjugated peptibody, was developed as a specific inhibitor with high avidity antagonizing both soluble and membrane-bound BAFF [43]. Nonetheless, neither atacicept nor blisibimod met clinical expectations in SLE treatment, highlighting the challenges within this field [20].

2.4. Ianalumab (VAY736)—mAb Directly Targeting B Cells

Ianalumab is a fully human mAb that specifically targets BAFF-R to lyse B-cells through ADCC and interrupt B-cell maturation, proliferation, and survival via blocking BAFF-mediated signaling [44]. It has been explored for potential therapeutic effects in various autoimmune conditions including MS, SLE, LN, and Sjögren’s disease, and more recently in patients with diffuse cutaneous system sclerosis. Subcutaneous administration of ianalumab in a randomized, double-blind, placebo-controlled study was well-tolerated and resulted in clinical improvements in Sjögren’s patients with statistically significant dose–response for overall disease activity [21].

2.5. Dazodalibep (HZN4920/AMG611)—HSA-Fusion Protein Antagonizing CD40L

Dazodalibep is a novel CD40 ligand (CD40L) antagonist. It is human serum albumin (HSA)-fusion protein possessing two Tn3 scaffolds [27], derived from the third fibronectin type III domain of human tenascin-C. Tn3 contains Ig-like folds, structurally analogous to antibody complementarity-determining regions (CDR). The Tn3 scaffolds of dazodalibep have been engineered to confer binding specificity to CD40L [28]. Lacking an Fc domain, dazodalibep does not lead to platelet aggregation or activation [28]. As an indirect and non-depleting B-cell modulator, dazodalibep blocks CD40L on T cells interacting with CD40-expressing B cells and disrupts the overactivation of the CD40 co-stimulatory pathway. Its clinical trials for several autoimmune diseases, such as Sjögren’s disease, kidney transplant rejection, and rheumatoid arthritis, are ongoing or have been completed. In addition to the positive data from the Phase 2 trial in patients with RA [26], the results from the Phase 2 trial for patients with Sjögren’s syndrome met the primary endpoint and led to the design of a Phase 3 program [45].

2.6. PRV-3279—Bispecific Antibody

PRV-3279 (formerly MGD010) is a humanized dual-affinity-retargeting (DART) bispecific molecule. This antibody targets B-cell surface proteins CD32B and CD79B simultaneously, developed by MacroGenics and licensed by Provention Bio. CD32B (FcγRIIB) is a low-affinity inhibitory receptor for IgG [46], while CD79B is part of the B cell receptor complex. Targeting CD79B alone has shown efficacy in treating autoimmune diseases in animal models [47]. Crosslinking CD32B and CD79B enhances downregulation of B-cell receptor signaling [48]. This non-depleting B-cell modulating bispecific drug aims to treat lupus and other autoimmune diseases and is currently in Phase 2a clinical trials for moderate-to-severe SLE patients (NCT05087628) [31].

3. Impact of Molecule Format on Manufacturability

While the most common type of molecule format used in B-cell targeting biologics is monoclonal antibody (mAb), other molecules with more diversified formats and enhanced features have been developed over the years (Figure 1). Many biologics were originally developed for cancer treatments and have since been repurposed for autoimmune diseases [10,17]. However, each new format presents its own set of challenges during the development and scale-up of the manufacturing process and some of them may need special considerations for manufacturability assessment and process development.

3.1. Molecule Format Selection and Process Development

Manufacturability is a critical consideration to ensure the ability to manufacture a product with the desired quality at an optimized cost. In the context of biologics discovery and development (Figure 2), manufacturability assessment should be performed at an early stage through collaboration between discovery and development teams; continued evaluation and optimization are iteratively needed to improve molecule format selection, molecule design and may extend through to the entire product lifecycle management. Such reiteration could lead to “next generation” molecule design [49]. The development and commercialization of next-generation anti-CD20 antibody therapies are typical examples of these efforts [7,33]. Platform approach using standard mAb processes can be a good starting point, but off-platform methods and further optimization are often needed to ensure high-quality and cost-effective manufacturing. For example, the production of afucosylated monoclonal antibodies may require the use of specialized host cells to achieve the desired product quality attributes.

3.1.1. Afucosylated mAb

Antibody glycosylation changes can significantly modulate its effector function [50]. When B-cell depletion is an intended effect, afucosylated glycoform is desirable for a therapeutic mAb, where afucosylation strongly increases IgG affinity to FcγRIIIa and thus ADCC activity [37,51,52]. Controlling glycoforms in manufacturing is challenging. Optimizing cell culture conditions can involve using small molecules to inhibit recombinant protein fucosylation [53]. However, a more efficient approach is using host cell lines with altered fucosylation processing (Table 2). These hosts may lack the ability to add fucose during glycosylation or have a shunted GDP-fucose pathway. As an example of FDA-approved afucosylated mAb drugs, UPLIZNA is produced by a proprietary FUT8−/− CHO cell line [36,37]. In addition to these CHO cell lines, other host cells for afucosylated mAb productions are also reported; rat hybridoma YB2/0 cells (ATCC® CRL1662™) intrinsically have lower levels of the FUT8 mRNA [54,55] and EB66 (derived from duck embryonic stem cells) have naturally reduced fucose content in the cells [52].

3.1.2. Fusion Protein

Since the approval of the etanercept for RA in 1998 [63], fusion proteins have emerged as one of the most used molecule formats in autoimmune disease treatments. However, their unique structures that are often derived from different cellular locations or cell types may lead to expression issues. Fusion protein heterogeneity in isoelectric point, charge densities, and hydrophobicity could further pose challenges in manufacturing. The linkers between individual modules and their orientation are typically designed based on functional requirements. However, these elements must undergo rigorous evaluation for structural stability and quality attributes to ensure manufacturability. This initial assessment is crucial for effectively minimizing challenges related to structural integrity. For example, O-glycosylation site elimination in linker regions was found to confer Fc fusion proteins with more homogenous glycans and less aggregative propensity [64]. Rearrangement of the disulfide bonding pattern was demonstrated to not only reduce aggregate formation but also improve pharmacokinetic properties [65].
Fusion protein glycoforms can be more complex than standard mAbs, necessitating optimization of medium compositions. Bioreactor conditions such as temperature shifts and harvest timing must also be evaluated for their impact on glycan attributes [66,67]. Medium optimization for glycosylation and sialylation can be achieved by adjusting key component concentrations [68,69] or adding specific additives [70,71]. Hydrocortisone and dexamethasone enhance Fc-fusion protein sialylation [70], while LongR3, an insulin-like growth factor-I analog, increases sialic acid content and reduces asialylated N-glycan species [71].
Fusion proteins may form aggregates during upstream cultivation and downstream purification. Disulfide bond-containing fusion protein aggregation can be minimized by balancing redox equivalents in media and culture conditions [72] or regulating glutathione reductase activity by glucocorticoid receptor agonists [73]. The pH dependency of fusion domain interactions and conformational stabilities, where a low pH may induce aggregate formation, would require downstream processing to avoid acidic elution from a capture column [74] and the use of low pH for virus inactivation.

3.1.3. Bispecific Antibody

Bispecifics can generally be manufactured using the same process as those for mAbs due to their structural similarities. However, variations in module formats can lead to differences in bispecifics. The manufacturing challenges include mispairing, high aggregation, and fragment formation, which necessitate early identification of sequence liabilities and the associated expression systems to ensure the correct assembly of different polypeptide chains [75]. Domain crossover, Fc heterodimerization, and common light chain strategies in protein engineering have been developed to design bispecifics with a lower tendency for mispairing and enhanced recombinant product stability.
A high-producer cell line that pairs the desired heterodimer is probably the key to allowing standard upstream and downstream processes to be utilized for bispecific production. For cell line development, titrations of separate gene constructs for transfection can increase the likelihood of correct protein assembly. Alternatively, different strengths of promoters should be used if all genes of interest are built into a single expression vector. Titer assay that measures the desired heterodimer form should be applied for rigorous clone screening.
As with standard mAb production, optimization of medium composition and culture conditions can affect product quality. To minimize aggregate formation and maximize the yield of a desired heterodimer, attention would particularly be paid to balancing redox reactions in media such as fine-tuning the concentrations of cysteine and metal ions [76]. In combination with closely monitoring overall quality attributes, temperature shifting is one of the most used strategies to improve process robustness and reduce product structural-related impurity formation [77]. To facilitate the removal of unwanted homodimers, the resin MabSelect SuRe™ pcc that has a small bead size and decreased binding avidity can be applied to the protein A matrix for high-resolution purification [78].

3.2. Scalability Considerations for Manufacturability Improvement

As part of manufacturability improvement, scalability assessment further ensures that the drug candidate, featuring the selected molecule format, is manufacturable by the newly developed cell line and process. Additionally, this new drug must be consistently producible across different scales with minimal parameter adjustments. This often includes tech transfer, facility fit analysis, and engineering runs, focusing on vessel geometry, mixing, and gassing. When lactate accumulation is higher and cell viability declines faster in large-scale cultures, optimizing agitation and oxygenation is usually the first step. Trace metal concentrations, particularly copper, may also need adjustment to prevent issues [79,80,81].
Despite significant efforts for process scaling-up, discussions on the scalability of cell lines for manufacturing are less common. Accelerated timelines for cell line development, driven by new technologies and automation, can lead to genetic instability and increased sensitivity to environmental stress [82], potentially inducing epigenetic modifications, lactate accumulation, and even cell death [81,83]. The addition of copper has been found to be effective in mitigating some of these issues but may not address all underlying causes.
Increased selection pressure during seed train expansion has been shown to improve production performance and scalability in CHO cultures without altering gene integration or copy number [84]. Lower lactate concentrations and enhanced re-maturation were observed, suggesting that re-cloning underperforming cell lines can enhance productivity and scalability.

4. Closing Remarks

B-cell-targeting biologics are innovative and successful new therapies for autoimmune diseases. While B-cell depletion therapy has shown efficacy in treating a range of autoimmune diseases, pan B-cell depletion can lead to broad immune suppression, increasing the risk of infections [85,86]. Further improvements in clinical efficacy and safety would therefore rely on continued efforts to discover new targets and to develop new molecule formats that specifically remove or inactivate pathogenic effector B cells while preserving regulatory B-cells for maintaining immune surveillance and minimizing adverse effects. While boosting potencies through avidity for deeper and more sustained effects, the bispecific format allows distinct targets to be simultaneously engaged and potentially increases the selectivity on certain populations of B-cells, such as autoantibodies-producing subpopulation, to avoid broad immune suppression. A bispecific format can also be applied in designing biologics that interact with two different cell types, such as B-cells and T-cells. T-cell mediated destruction of pathogenetic B-cells offers a different mode of action compared with the pan B-cell depletion through complement-dependent cytotoxicity and antibody-dependent cellular cytotoxicity; simultaneously targeting B-cells and engaging T-cells can help reset the immune system [17]. Product manufacturability needs to be evaluated as early as possible and iteratively with the inputs from process development teams. Selecting the right expression system and cell line based on the molecule format-specific needs provides an upfront advantage for maximizing titer while maintaining product quality attributes, which in turn simplifies later purification and formulation steps. Platform approach using standard mAb processes can be a good starting point, but it should be complemented with a well-characterized and molecule format-specific toolbox for further optimization to ensure high quality and cost-effective manufacturing. The toolbox contains various levelers of medium compositions including additives, resin selections, and in-process controls, but more importantly, it is empowered by deep mechanistic understanding. New technologies are on the horizon. We need continued learning, innovation, and exploration.

Author Contributions

Conceptualization, Y.Q.; literature search and analysis, Y.Q. and T.I.M.; writing—original draft preparation, Y.Q.; writing—review and editing, Y.Q. and T.I.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Acknowledgments

The authors would like to thank Natalia Gomez for her continued management support, manuscript review, and valuable suggestions.

Conflicts of Interest

Y.Q. and T.M. are employees of Amgen Inc. and own stock. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Kurosaki, T. B-lymphocyte biology. Immunol. Rev. 2010, 237, 5–9. [Google Scholar] [CrossRef]
  2. Townsend, M.J.; Monroe, J.G.; Chan, A.C. B-cell targeted therapies in human autoimmune diseases: An updated perspective. Immunol. Rev. 2010, 237, 264–283. [Google Scholar] [CrossRef]
  3. Barnas, J.L.; Looney, R.J.; Anolik, J.H. B cell targeted therapies in autoimmune disease. Curr. Opin. Immunol. 2019, 61, 92–99. [Google Scholar] [CrossRef] [PubMed]
  4. Frampton, J.E. Inebilizumab: First Approval. Drugs 2020, 80, 1259–1264. [Google Scholar] [CrossRef] [PubMed]
  5. Stensland, Z.C.; Cambier, J.C.; Smith, M.J. Therapeutic Targeting of Autoreactive B Cells: Why, How, and When? Biomedicines 2021, 9, 83. [Google Scholar] [CrossRef] [PubMed]
  6. Merino-Vico, A.; Frazzei, G.; van Hamburg, J.P.; Tas, S.W. Targeting B cells and plasma cells in autoimmune diseases: From established treatments to novel therapeutic approaches. Eur. J. Immunol. 2023, 53, e2149675. [Google Scholar] [CrossRef]
  7. Du, F.H.; Mills, E.A.; Mao-Draayer, Y. Next-generation anti-CD20 monoclonal antibodies in autoimmune disease treatment. Auto. Immun. Highlights 2017, 8, 12. [Google Scholar] [CrossRef] [PubMed]
  8. Huda, R. New Approaches to Targeting B Cells for Myasthenia Gravis Therapy. Front. Immunol. 2020, 11, 240. [Google Scholar] [CrossRef]
  9. Hofmann, K.; Clauder, A.K.; Manz, R.A. Targeting B Cells and Plasma Cells in Autoimmune Diseases. Front. Immunol. 2018, 9, 835. [Google Scholar] [CrossRef]
  10. Gurcan, H.M.; Keskin, D.B.; Stern, J.N.; Nitzberg, M.A.; Shekhani, H.; Ahmed, A.R. A review of the current use of rituximab in autoimmune diseases. Int. Immunopharmacol. 2009, 9, 10–25. [Google Scholar] [CrossRef]
  11. Kanatas, P.; Stouras, I.; Stefanis, L.; Stathopoulos, P. B-Cell-Directed Therapies: A New Era in Multiple Sclerosis Treatment. Can. J. Neurol. Sci. 2022, 20, 355–364. [Google Scholar] [CrossRef]
  12. Rubbert-Roth, A. TRU-015, a fusion protein derived from an anti-CD20 antibody, for the treatment of rheumatoid arthritis. Curr. Opin. Mol. Ther. 2010, 12, 115–123. [Google Scholar] [PubMed]
  13. Robinson, W.H.; Fiorentino, D.; Chung, L.; Moreland, L.W.; Deodhar, M.; Harler, M.B.; Saulsbery, C.; Kunder, R. Cutting-edge approaches to B-cell depletion in autoimmune diseases. Front. Immunol. 2024, 15, 1454747. [Google Scholar] [CrossRef]
  14. Stone, J.H.; Khosroshahi, A.; Zhang, W.; Della Torre, E.; Okazaki, K.; Tanaka, Y.; Lohr, J.M.; Schleinitz, N.; Dong, L.; Umehara, H.; et al. Inebilizumab for Treatment of IgG4-Related Disease. N. Engl. J. Med. 2022, 132, 102873. [Google Scholar] [CrossRef]
  15. Arbitman, L.; Furie, R.; Vashistha, H. B cell-targeted therapies in systemic lupus erythematosus. J. Autoimmun. 2022, 132, 102873. [Google Scholar] [CrossRef] [PubMed]
  16. Subklewe, M.; Magno, G.; Gebhardt, C.; Bucklein, V.; Szelinski, F.; Arevalo, H.J.R.; Hanel, G.; Dorner, T.; Zugmaier, G.; von Bergwelt-Baildon, M.; et al. Application of blinatumomab, a bispecific anti-CD3/CD19 T-cell engager, in treating severe systemic sclerosis: A case study. Eur. J. Cancer 2024, 204, 114071. [Google Scholar] [CrossRef]
  17. Zugmaier, G.; Klinger, M.; Subklewe, M.; Zaman, F.; Locatelli, F. B-Cell-Depleting Immune Therapies as Potential New Treatment Options for Systemic Sclerosis. Sclerosis 2025, 3, 5. [Google Scholar] [CrossRef]
  18. Li, J.; Li, M.; Wu, D.; Zhou, J.; Leung, S.O.; Zhang, F. SM03, an anti-human CD22 monoclonal antibody, for active rheumatoid arthritis: A phase II, randomized, double-blind, placebo-controlled study. Rheumatology 2022, 61, 1841–1848. [Google Scholar] [CrossRef]
  19. Geh, D.; Gordon, C. Epratuzumab for the treatment of systemic lupus erythematosus. Expert. Rev. Clin. Immunol. 2018, 14, 245–258. [Google Scholar] [CrossRef]
  20. Runkel, L.; Stacey, J. Lupus clinical development: Will belimumab’s approval catalyse a new paradigm for SLE drug development? Expert. Opin. Biol. Ther. 2014, 14, 491–501. [Google Scholar] [CrossRef]
  21. Bowman, S.J.; Fox, R.; Dorner, T.; Mariette, X.; Papas, A.; Grader-Beck, T.; Fisher, B.A.; Barcelos, F.; De Vita, S.; Schulze-Koops, H.; et al. Safety and efficacy of subcutaneous ianalumab (VAY736) in patients with primary Sjogren’s syndrome: A randomised, double-blind, placebo-controlled, phase 2b dose-finding trial. Lancet 2022, 399, 161–171. [Google Scholar] [CrossRef] [PubMed]
  22. Furie, R.A.; Bruce, I.N.; Dorner, T.; Leon, M.G.; Leszczynski, P.; Urowitz, M.; Haier, B.; Jimenez, T.; Brittain, C.; Liu, J.; et al. Phase 2, randomized, placebo-controlled trial of dapirolizumab pegol in patients with moderate-to-severe active systemic lupus erythematosus. Rheumatology 2021, 60, 5397–5407. [Google Scholar] [CrossRef]
  23. Kahaly, G.J.; Stan, M.N.; Frommer, L.; Gergely, P.; Colin, L.; Amer, A.; Schuhmann, I.; Espie, P.; Rush, J.S.; Basson, C.; et al. A Novel Anti-CD40 Monoclonal Antibody, Iscalimab, for Control of Graves Hyperthyroidism-A Proof-of-Concept Trial. J. Clin. Endocrinol. Metab. 2020, 105, 696–704. [Google Scholar] [CrossRef]
  24. Fisher, B.A.; Szanto, A.; Ng, W.-F.; Bombardieri, M.; Gergely, P. Assessment of the anti-CD40 antibody iscalimab in patients with primary Sjögren’s syndrome: A multicentre, randomised, double-blind, placebo-controlled, proof-of-concept study. Lancet Rheumatol. 2020, 2, 11. [Google Scholar] [CrossRef] [PubMed]
  25. Visvanathan, S.; Daniluk, S.; Ptaszynski, R.; Muller-Ladner, U.; Ramanujam, M.; Rosenstock, B.; Eleftheraki, A.G.; Vinisko, R.; Petrikova, A.; Kellner, H.; et al. Effects of BI 655064, an antagonistic anti-CD40 antibody, on clinical and biomarker variables in patients with active rheumatoid arthritis: A randomised, double-blind, placebo-controlled, phase IIa study. Ann. Rheum. Dis. 2019, 78, 754–760. [Google Scholar] [CrossRef]
  26. Kivitz, A. A Phase 2, Randomized, Double-Blind, Placebo-Controlled, Mechanistic Insight and Dosage Optimization Study of the Efficacy and Safety of Dazodalibep (VIB4920/HZN4920) in Patients with Rheumatoid Arthritis Having Inadequate Response to Conventional/Biological DMARDs. Arthritis Rheumatol. 2022, 74, 1045–1048. [Google Scholar]
  27. Oganesyan, V.; Ferguson, A.; Grinberg, L.; Wang, L.; Phipps, S.; Chacko, B.; Drabic, S.; Thisted, T.; Baca, M. Fibronectin type III domains engineered to bind CD40L: Cloning, expression, purification, crystallization and preliminary X-ray diffraction analysis of two complexes. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 2013, 69, 1045–1048. [Google Scholar] [CrossRef]
  28. Karnell, J.L.; Rieder, S.A.; Ettinger, R.; Kolbeck, R. Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond. Adv. Drug Deliv. Rev. 2019, 141, 92–103. [Google Scholar] [CrossRef]
  29. Dhillon, S. Telitacicept: First Approval. Drugs 2021, 81, 1671–1675. [Google Scholar] [CrossRef]
  30. Bracewell, C.; Isaacs, J.D.; Emery, P.; Ng, W.F. Atacicept, a novel B cell-targeting biological therapy for the treatment of rheumatoid arthritis. Expert. Opin. Biol. Ther. 2009, 9, 909–919. [Google Scholar] [CrossRef]
  31. Dunford, P.; Comer, G.; Raymond, R.; Jung, D.; Moore, P.; Leon, F.; Merrill, J.T. PREVAIL 1: A Multiple Ascending Dose Study in Normal Healthy Volunteers of PRV-3279, a Novel Bispecific DART Molecule Targeting CD32B and CD79B on B Cells, with Potential for Treatment of SLE. Arthritis Rheumatol. 2020, 72, 13–25. [Google Scholar]
  32. Robak, T.; Robak, E. New anti-CD20 monoclonal antibodies for the treatment of B-cell lymphoid malignancies. BioDrugs 2011, 25, 13–25. [Google Scholar] [CrossRef] [PubMed]
  33. Frampton, J.E. Ocrelizumab: First Global Approval. Drugs 2017, 77, 1035–1041. [Google Scholar] [CrossRef] [PubMed]
  34. McCool, R.; Wilson, K.; Arber, M.; Fleetwood, K.; Toupin, S.; Thom, H.; Bennett, I.; Edwards, S. Systematic review and network meta-analysis comparing ocrelizumab with other treatments for relapsing multiple sclerosis. Mult. Scler. Relat. Disord. 2019, 29, 55–61. [Google Scholar] [CrossRef]
  35. Hauser, S.L.; Bar-Or, A.; Cohen, J.A.; Comi, G.; Correale, J.; Coyle, P.K.; Cross, A.H.; de Seze, J.; Leppert, D.; Montalban, X.; et al. Ofatumumab versus Teriflunomide in Multiple Sclerosis. N. Engl. J. Med. 2020, 383, 546–557. [Google Scholar] [CrossRef] [PubMed]
  36. Herbst, R.; Wang, Y.; Gallagher, S.; Mittereder, N.; Kuta, E.; Damschroder, M.; Woods, R.; Rowe, D.C.; Cheng, L.; Cook, K.; et al. B-cell depletion in vitro and in vivo with an afucosylated anti-CD19 antibody. J. Pharmacol. Exp. Ther. 2010, 335, 213–222. [Google Scholar] [CrossRef]
  37. Gallagher, S.; Turman, S.; Yusuf, I.; Akhgar, A.; Wu, Y.; Roskos, L.K.; Herbst, R.; Wang, Y. Pharmacological profile of MEDI-551, a novel anti-CD19 antibody, in human CD19 transgenic mice. Int. Immunopharmacol. 2016, 36, 205–212. [Google Scholar] [CrossRef]
  38. Nie, T.; Blair, H.A. Inebilizumab: A Review in Neuromyelitis Optica Spectrum Disorder. CNS Drugs 2022, 36, 1133–1141. [Google Scholar] [CrossRef]
  39. Rensel, M.; Zabeti, A.; Mealy, M.A.; Cimbora, D.; She, D.; Drappa, J.; Katz, E. Long-term efficacy and safety of inebilizumab in neuromyelitis optica spectrum disorder: Analysis of aquaporin-4-immunoglobulin G-seropositive participants taking inebilizumab for ⩾4 years in the N-MOmentum trial. Mult. Scler. 2022, 28, 925–932. [Google Scholar] [CrossRef]
  40. Zhang, Y.; Tian, J.; Xiao, F.; Zheng, L.; Zhu, X.; Wu, L.; Zhao, C.; Wang, S.; Rui, K.; Zou, H.; et al. B cell-activating factor and its targeted therapy in autoimmune diseases. Cytokine Growth Factor. Rev. 2022, 64, 57–70. [Google Scholar] [CrossRef]
  41. Baker, D.; Pryce, G.; James, L.K.; Schmierer, K.; Giovannoni, G. Failed B cell survival factor trials support the importance of memory B cells in multiple sclerosis. Eur. J. Neurol. 2020, 27, 221–228. [Google Scholar] [CrossRef] [PubMed]
  42. Kramer, J.; Wiendl, H. What Have Failed, Interrupted, and Withdrawn Antibody Therapies in Multiple Sclerosis Taught Us? Neurotherapeutics 2022, 19, 785–807. [Google Scholar] [CrossRef]
  43. Lenert, A.; Niewold, T.B.; Lenert, P. Spotlight on blisibimod and its potential in the treatment of systemic lupus erythematosus: Evidence to date. Drug Des. Devel Ther. 2017, 11, 747–757. [Google Scholar] [CrossRef] [PubMed]
  44. Cuker, A.; Al-Samkari, H.; Barcellini, W.; Cooper, N.; Ghanima, W.; Michel, M.; Wong, R.S.; Zaja, F.; Zhang, F.; Urban, P.; et al. Ianalumab, a Novel Anti-B-Cell Activating Factor (BAFF) Receptor (BAFF-R) Monoclonal Antibody (mAb) in Development for Immune Thrombocytopenia (ITP) and Warm Autoimmune Hemolytic Anemia (wAIHA), Has Demonstrated a Favorable Safety Profile in Sjögren’s Syndrome (SjS), Systemic Lupus Erythematosus (SLE) and Chronic Lymphocytic Leukemia (CLL). Blood 2023, 142, 5427. [Google Scholar] [CrossRef]
  45. St Clair, E.W.; Baer, A.N.; Ng, W.F.; Noaiseh, G.; Baldini, C.; Tarrant, T.K.; Papas, A.; Devauchelle-Pensec, V.; Wang, L.; Xu, W.; et al. CD40 ligand antagonist dazodalibep in Sjogren’s disease: A randomized, double-blinded, placebo-controlled, phase 2 trial. Nat. Med. 2024, 30, 1583–1592. [Google Scholar] [CrossRef]
  46. Amigorena, S.; Bonnerot, C.; Drake, J.R.; Choquet, D.; Hunziker, W.; Guillet, J.G.; Webster, P.; Sautes, C.; Mellman, I.; Fridman, W.H. Cytoplasmic domain heterogeneity and functions of IgG Fc receptors in B lymphocytes. Science 1992, 256, 1808–1812. [Google Scholar] [CrossRef]
  47. Renner, K.; Neumayer, S.; Talke, Y.; Buchtler, S.; Schmidbauer, K.; Nimmerjahn, F.; Lux, A.; Winter, F.; Salewski, J.N.; Mack, M. B-cell modulation with anti-CD79b antibodies ameliorates experimental autoimmune encephalitis in mice. Eur. J. Immunol. 2022, 52, 656–668. [Google Scholar] [CrossRef]
  48. Veri, M.C.; Burke, S.; Huang, L.; Li, H.; Gorlatov, S.; Tuaillon, N.; Rainey, G.J.; Ciccarone, V.; Zhang, T.; Shah, K.; et al. Therapeutic control of B cell activation via recruitment of Fcgamma receptor IIb (CD32B) inhibitory function with a novel bispecific antibody scaffold. Arthritis Rheum. 2010, 62, 1933–1943. [Google Scholar] [CrossRef]
  49. Chen, Z.; Qian, Y.; Song, Y.; Xu, X.; Tao, L.; Mussa, N.; Ghose, S.; Li, Z.J. Design of next-generation therapeutic IgG4 with improved manufacturability and bioanalytical characteristics. MAbs 2020, 12, 1829338. [Google Scholar] [CrossRef]
  50. Thomann, M.; Reckermann, K.; Reusch, D.; Prasser, J.; Tejada, M.L. Fc-galactosylation modulates antibody-dependent cellular cytotoxicity of therapeutic antibodies. Mol. Immunol. 2016, 73, 69–75. [Google Scholar] [CrossRef]
  51. Shields, R.L.; Lai, J.; Keck, R.; O’Connell, L.Y.; Hong, K.; Meng, Y.G.; Weikert, S.H.; Presta, L.G. Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J. Biol. Chem. 2002, 277, 26733–26740. [Google Scholar] [CrossRef] [PubMed]
  52. Olivier, S.; Jacoby, M.; Brillon, C.; Bouletreau, S.; Mollet, T.; Nerriere, O.; Angel, A.; Danet, S.; Souttou, B.; Guehenneux, F.; et al. EB66 cell line, a duck embryonic stem cell-derived substrate for the industrial production of therapeutic monoclonal antibodies with enhanced ADCC activity. MAbs 2010, 2, 405–415. [Google Scholar] [CrossRef]
  53. Okeley, N.M.; Alley, S.C.; Anderson, M.E.; Boursalian, T.E.; Burke, P.J.; Emmerton, K.M.; Jeffrey, S.C.; Klussman, K.; Law, C.L.; Sussman, D.; et al. Development of orally active inhibitors of protein and cellular fucosylation. Proc. Natl. Acad. Sci. USA 2013, 110, 5404–5409. [Google Scholar] [CrossRef] [PubMed]
  54. Kilmartin, J.V.; Wright, B.; Milstein, C. Rat monoclonal antitubulin antibodies derived by using a new nonsecreting rat cell line. J. Cell Biol. 1982, 93, 576–582. [Google Scholar] [CrossRef]
  55. Shinkawa, T.; Nakamura, K.; Yamane, N.; Shoji-Hosaka, E.; Kanda, Y.; Sakurada, M.; Uchida, K.; Anazawa, H.; Satoh, M.; Yamasaki, M.; et al. The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J. Biol. Chem. 2003, 278, 3466–3473. [Google Scholar] [CrossRef]
  56. Ripka, J.; Adamany, A.; Stanley, P. Two Chinese hamster ovary glycosylation mutants affected in the conversion of GDP-mannose to GDP-fucose. Arch. Biochem. Biophys. 1986, 249, 533–545. [Google Scholar] [CrossRef] [PubMed]
  57. Yamane-Ohnuki, N.; Kinoshita, S.; Inoue-Urakubo, M.; Kusunoki, M.; Iida, S.; Nakano, R.; Wakitani, M.; Niwa, R.; Sakurada, M.; Uchida, K.; et al. Establishment of FUT8 knockout Chinese hamster ovary cells: An ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity. Biotechnol. Bioeng. 2004, 87, 614–622. [Google Scholar] [CrossRef]
  58. Malphettes, L.; Freyvert, Y.; Chang, J.; Liu, P.Q.; Chan, E.; Miller, J.C.; Zhou, Z.; Nguyen, T.; Tsai, C.; Snowden, A.W.; et al. Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies. Biotechnol. Bioeng. 2010, 106, 774–783. [Google Scholar] [CrossRef]
  59. Chan, K.F.; Shahreel, W.; Wan, C.; Teo, G.; Hayati, N.; Tay, S.J.; Tong, W.H.; Yang, Y.; Rudd, P.M.; Zhang, P.; et al. Inactivation of GDP-fucose transporter gene (Slc35c1) in CHO cells by ZFNs, TALENs and CRISPR-Cas9 for production of fucose-free antibodies. Biotechnol. J. 2016, 11, 399–414. [Google Scholar] [CrossRef]
  60. von Horsten, H.H.; Ogorek, C.; Blanchard, V.; Demmler, C.; Giese, C.; Winkler, K.; Kaup, M.; Berger, M.; Jordan, I.; Sandig, V. Production of non-fucosylated antibodies by co-expression of heterologous GDP-6-deoxy-D-lyxo-4-hexulose reductase. Glycobiology 2010, 20, 1607–1618. [Google Scholar] [CrossRef]
  61. Ferrara, C.; Brunker, P.; Suter, T.; Moser, S.; Puntener, U.; Umana, P. Modulation of therapeutic antibody effector functions by glycosylation engineering: Influence of Golgi enzyme localization domain and co-expression of heterologous beta1, 4-N-acetylglucosaminyltransferase III and Golgi alpha-mannosidase II. Biotechnol. Bioeng. 2006, 93, 851–861. [Google Scholar] [CrossRef] [PubMed]
  62. Kanda, Y.; Imai-Nishiya, H.; Kuni-Kamochi, R.; Mori, K.; Inoue, M.; Kitajima-Miyama, K.; Okazaki, A.; Iida, S.; Shitara, K.; Satoh, M. Establishment of a GDP-mannose 4,6-dehydratase (GMD) knockout host cell line: A new strategy for generating completely non-fucosylated recombinant therapeutics. J. Biotechnol. 2007, 130, 300–310. [Google Scholar] [CrossRef]
  63. Nanda, S.; Bathon, J.M. Etanercept: A clinical review of current and emerging indications. Expert. Opin. Pharmacother. 2004, 5, 1175–1186. [Google Scholar] [CrossRef]
  64. Song, Y.; Qian, Y.; Huang, Z.; Khattak, S.F.; Li, Z.J. Computational insights into O-glycosylation in a CTLA4 Fc-fusion protein linker and its impact on protein quality attributes. Comput. Struct. Biotechnol. J. 2020, 18, 3925–3935. [Google Scholar] [CrossRef] [PubMed]
  65. Way, J.C.; Lauder, S.; Brunkhorst, B.; Kong, S.M.; Qi, A.; Webster, G.; Campbell, I.; McKenzie, S.; Lan, Y.; Marelli, B.; et al. Improvement of Fc-erythropoietin structure and pharmacokinetics by modification at a disulfide bond. Protein Eng. Des. Sel. 2005, 18, 111–118. [Google Scholar] [CrossRef]
  66. Trummer, E.; Fauland, K.; Seidinger, S.; Schriebl, K.; Lattenmayer, C.; Kunert, R.; Vorauer-Uhl, K.; Weik, R.; Borth, N.; Katinger, H.; et al. Process parameter shifting: Part I. Effect of DOT, pH, and temperature on the performance of Epo-Fc expressing CHO cells cultivated in controlled batch bioreactors. Biotechnol. Bioeng. 2006, 94, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
  67. Hossler, P.; Khattak, S.F.; Li, Z.J. Optimal and consistent protein glycosylation in mammalian cell culture. Glycobiology 2009, 19, 936–949. [Google Scholar] [CrossRef]
  68. Ha, T.K.; Lee, G.M. Effect of glutamine substitution by TCA cycle intermediates on the production and sialylation of Fc-fusion protein in Chinese hamster ovary cell culture. J. Biotechnol. 2014, 180, 23–29. [Google Scholar] [CrossRef]
  69. Taschwer, M.; Hackl, M.; Hernandez Bort, J.A.; Leitner, C.; Kumar, N.; Puc, U.; Grass, J.; Papst, M.; Kunert, R.; Altmann, F.; et al. Growth, productivity and protein glycosylation in a CHO EpoFc producer cell line adapted to glutamine-free growth. J. Biotechnol. 2012, 157, 295–303. [Google Scholar] [CrossRef]
  70. Jing, Y.; Qian, Y.; Li, Z.J. Sialylation enhancement of CTLA4-Ig fusion protein in Chinese hamster ovary cells by dexamethasone. Biotechnol. Bioeng. 2010, 107, 488–496. [Google Scholar] [CrossRef]
  71. Qian, Y.; Lewis, A.M.; Sidnam, S.M.; Bergeron, A.; Abu-Absi, N.R.; Vaidyanathan, N.; Deresienski, A.; Qian, N.-X.; Borys, M.C.; Li, Z.J. LongR3 enhances Fc-fusion protein N-linked glycosylation while improving protein productivity in an industrial CHO cell line. Process Biochem. 2017, 53, 9. [Google Scholar] [CrossRef]
  72. Ying, J.; Borys, B.C.; Samiksha, N.; Egan, S.; Qian, Y.; Pan, S.-H.; Li, Z.J. Identification of cell culture conditions to control protein aggregation of IgG fusion proteins expressed in Chinese hamster ovary cells. Process Biochem. 2012, 47, 6. [Google Scholar] [CrossRef]
  73. Qian, Y.; Jing, Y.; Li, Z.J. Glucocorticoid receptor-mediated reduction of IgG-fusion protein aggregation in Chinese hamster ovary cells. Biotechnol. Prog. 2010, 26, 1417–1423. [Google Scholar] [CrossRef] [PubMed]
  74. Shukla, A.A.; Gupta, P.; Han, X. Protein aggregation kinetics during Protein A chromatography. Case study for an Fc fusion protein. J. Chromatogr. A 2007, 1171, 22–28. [Google Scholar] [CrossRef]
  75. Wang, Q.; Chen, Y.; Park, J.; Liu, X.; Hu, Y.; Wang, T.; McFarland, K.; Betenbaugh, M.J. Design and Production of Bispecific Antibodies. Antibodies 2019, 8, 998–1008. [Google Scholar] [CrossRef] [PubMed]
  76. Purdie, J.L.; Kowle, R.L.; Langland, A.L.; Patel, C.N.; Ouyang, A.; Olson, D.J. Cell culture media impact on drug product solution stability. Biotechnol. Prog. 2016, 32, 998–1008. [Google Scholar] [CrossRef]
  77. Gomez, N.; Wieczorek, A.; Lu, F.; Bruno, R.; Diaz, L.; Agrawal, N.J.; Daris, K. Culture temperature modulates half antibody and aggregate formation in a Chinese hamster ovary cell line expressing a bispecific antibody. Biotechnol. Bioeng. 2018, 115, 2930–2940. [Google Scholar] [CrossRef]
  78. Tustian, A.D.; Laurin, L.; Ihre, H.; Tran, T.; Stairs, R.; Bak, H. Development of a novel affinity chromatography resin for platform purification of bispecific antibodies with modified protein a binding avidity. Biotechnol. Prog. 2018, 34, 650–658. [Google Scholar] [CrossRef]
  79. Brantley, T.; Moore, B.; Grinnell, C.; Khattak, S. Investigating trace metal precipitation in highly concentrated cell culture media with Pourbaix diagrams. Biotechnol. Bioeng. 2021, 118, 3888–3897. [Google Scholar] [CrossRef]
  80. Qian, Y.; Khattak, S.F.; Xing, Z.; He, A.; Kayne, P.S.; Qian, N.X.; Pan, S.H.; Li, Z.J. Cell culture and gene transcription effects of copper sulfate on Chinese hamster ovary cells. Biotechnol. Prog. 2011, 27, 1190–1194. [Google Scholar] [CrossRef]
  81. Qian, Y.; Xing, Z.; Lee, S.; Mackin, N.A.; He, A.; Kayne, P.S.; He, Q.; Qian, N.X.; Li, Z.J. Hypoxia influences protein transport and epigenetic repression of CHO cell cultures in shake flasks. Biotechnol. J. 2014, 9, 1413–1424. [Google Scholar] [CrossRef] [PubMed]
  82. Qian, Y.; Sowa, S.W.; Aron, K.L.; Xu, P.; Langsdorf, E.; Warrack, B.; Aranibar, N.; Tremml, G.; Xu, J.; Duncan, M.; et al. New insights into genetic instability of an industrial CHO cell line by orthogonal omics. Biochem. Eng. J. 2020, 164, 12. [Google Scholar] [CrossRef]
  83. Qian, Y.; Rehmann, M.S.; Qian, N.X.; He, A.; Borys, M.C.; Kayne, P.S.; Li, Z.J. Hypoxia and transforming growth factor-beta1 pathway activation promote Chinese Hamster Ovary cell aggregation. Biotechnol. Bioeng. 2018, 115, 1051–1061. [Google Scholar] [CrossRef] [PubMed]
  84. Tian, J.; He, Q.; Oliveira, C.; Qian, Y.; Egan, S.; Xu, J.; Qian, N.X.; Langsdorf, E.; Warrack, B.; Aranibar, N.; et al. Increased MSX level improves biological productivity and production stability in multiple recombinant GS CHO cell lines. Eng. Life Sci. 2020, 20, 112–125. [Google Scholar] [CrossRef] [PubMed]
  85. Luna, G.; Alping, P.; Burman, J.; Fink, K.; Fogdell-Hahn, A.; Gunnarsson, M.; Hillert, J.; Langer-Gould, A.; Lycke, J.; Nilsson, P.; et al. Infection Risks Among Patients with Multiple Sclerosis Treated With Fingolimod, Natalizumab, Rituximab, and Injectable Therapies. JAMA Neurol. 2020, 77, 184–191. [Google Scholar] [CrossRef]
  86. Safavi, F.; Nourbakhsh, B.; Azimi, A.R. B-cell depleting therapies may affect susceptibility to acute respiratory illness among patients with multiple sclerosis during the early COVID-19 epidemic in Iran. Mult. Scler. Relat. Disord. 2020, 43, 102195. [Google Scholar] [CrossRef]
Figure 1. Trend in molecule formats of B-cell-targeting biologics launched or clinically trialed for autoimmune diseases. With a better understanding of the field and technological advancement, more diversified formats and enhanced features have been developed with time. Many molecules have been proven successful in oncology and are being introduced for autoimmune disease treatment [10,17]. Approximate timeline in the diagram is for the time when those biologics entered the clinical trials for autoimmune indications. ❶ = rituximab, SM03; ❷ = ublituximab; ❸ = veltuzumab, epratuzumab; BI 655064; ❹ = ocrelizumab, inebilizumab; ❺ = ofatumumab, daratumumab, belimumab, ianalumab; ❻ = iscalimab; ❼ = telitacicept, atacicept; ❽ = TRU-015; ❾ = dapirolizumab pegol; ❿ = dazodalibep; ⓫ = PRV-3279, mosunetuzumab, obexelimab, blinatumomab; ⓬ = imvotamab; ⓭ = PIT565.
Figure 1. Trend in molecule formats of B-cell-targeting biologics launched or clinically trialed for autoimmune diseases. With a better understanding of the field and technological advancement, more diversified formats and enhanced features have been developed with time. Many molecules have been proven successful in oncology and are being introduced for autoimmune disease treatment [10,17]. Approximate timeline in the diagram is for the time when those biologics entered the clinical trials for autoimmune indications. ❶ = rituximab, SM03; ❷ = ublituximab; ❸ = veltuzumab, epratuzumab; BI 655064; ❹ = ocrelizumab, inebilizumab; ❺ = ofatumumab, daratumumab, belimumab, ianalumab; ❻ = iscalimab; ❼ = telitacicept, atacicept; ❽ = TRU-015; ❾ = dapirolizumab pegol; ❿ = dazodalibep; ⓫ = PRV-3279, mosunetuzumab, obexelimab, blinatumomab; ⓬ = imvotamab; ⓭ = PIT565.
Pharmaceutics 17 00495 g001
Figure 2. General workflow for advancing biologics development from discovery through to manufacturing. Different molecule formats may have different impacts on process development and manufacturing. Continued assessment and optimization may iteratively be needed for the development of biologics with a complex format such as bispecifics.
Figure 2. General workflow for advancing biologics development from discovery through to manufacturing. Different molecule formats may have different impacts on process development and manufacturing. Continued assessment and optimization may iteratively be needed for the development of biologics with a complex format such as bispecifics.
Pharmaceutics 17 00495 g002
Table 1. B-cell targeted biologics in autoimmune diseases and their molecule formats and features.
Table 1. B-cell targeted biologics in autoimmune diseases and their molecule formats and features.
TargetDrug NameMolecule Format/FeaturesAutoimmune IndicationsReferences
CD20RituximabChimeric murine-human IgG1k mAb/targeting CD20 on pro-B cells and all mature B cells, but not long-lived plasma or plasmablast cells.Approved: RA, GPA, MPA, PV
Clinical trials: ITP, MG
[10]
Ocrelizumab Humanized mAb/with afucosylated glycoforms enhancing ADCCApproved: RRMS and PPMS[11]
UblituximabChimeric murine-human IgG mAb/with low-fucosylated glycoforms enhancing ADCCApproved: RRMS, CIS, SPMS[11]
OfatumumabFully human monoclonal antibody/first B-cell-targeting therapy that is intended for self-administration at home Approved: RRMS, CIS, SPMS
Clinical trial: RA
[11]
VeltuzumabHumanized mAb/epratuzumab framework and rituximab CDRsFDA granted orphan status designation for ITP and pemphigus
Clinical trial: RA
[7]
TRU-015Fully human IgG fusion protein/a single-chain Fv specific for CD20 linked to human IgG1 hinge, CH2, and CH3 domains but devoid of CH1 and CL domainsClinical trials: active seropositive RA on a stable background of methotrexate[12]
MosunetuzumabBispecific antibody/IgG, anti-CD20 and anti-CD3Clinical trials:
SLE
[13]
ImvotamabBispecific antibody/IgM, anti-CD20 and anti-CD3Clinical trials:
RA, SLE
[13]
CD19InebilizumabHumanized IgG1k mAb/with afucosylated glycoforms enhancing ADCCApproved: NMOSD with AQP4-IgG+
Clinical trials: GM, IgG4-RD
[4,14]
NCT04524273
ObexelimabBispecific antibody/simultaneously binds CD19 and FcγRIIb, resulting in down-regulation of B cell activityClinical trials:
GM; IgG4-RD
[15]
BlinatumomabBispecific antibody/anti-CD19 and anti-CD3Clinical trials:
RA, system sclerosis
[16,17]
PIT565Trispecific antibody/anti-CD19, anti-CD3, and anti-CD2 Clinical trials:
SLE
NCT06335979
CD22SM03Chimeric murine-human mAb/targeting the extracellular portion of CD22Clinical trials: SLE, RA[18]
EpratuzumabHumanized mAb/targeting CD22 with modest ADCC activityClinical trials: SLE[19]
CD38DaratumumabFully human mAb/targeting CD38 on long-lived plasma cellsClinical trials: SLE[15]
BAFF/BAFF-RBelimumabFully human mAb/neutralizing biologically active soluble form of BAFFApproved: SLE and lupus nephritis[20]
Ianalumab
(VAY736)
Fully human mAb/antagonizing BAFF-RClinical trials: MS, SLE, Sjögren’s syndrome, Diffuse Cutaneous Systemic Sclerosis[21]
CD40/CD40LDapirolizumab pegolFab/polyethylene glycol-conjugated, anti-CD40L, lacking the Fc-portion to avoid platelet activationClinical trials: SLE[22]
Iscalimab (CFZ533)Fully human mAb/Fc-silenced, antagonizing CD40Clinical trials: Graves disease (GD); Sjögren’s syndrome[23,24]
BI 655064Humanized mAb/anti-CD40 blocking CD40-CD40L interactionClinical trials: RA[25]
Dazodalibep
(AMG611, HZN-4920)
Ig-like scaffold-HSA fusion protein/Tn3 scaffolds derived from the 3rd fibronectin type III domain of human tenascin-C, structurally analogous to antibody CDRs and functionally blocking CD40-CD40L interactionClinical trials: RA, Sjögren’s syndrome [26,27,28]
BAFF/APRILTelitaciceptFc fusion protein/fused with extracellular domain (amino acids 13-118) of TACI binding to and neutralizing BAFF and APRILApproved:
SLE (in China)
Clinical trials:
IgA nephropathy, MS, RA, MG, Sjögren’s syndrome
[29]
AtaciceptFc fusion protein/fused with extracellular domain (amino acids 30-110) of TACI binding to and neutralizing BAFF and APRILClinical trials: SLE, RA, IgA nephropathy[30]
CD32B/CD79BPRV-3279
(MGD010)
Bispecific antibody/simultaneously targeting B-cell surface proteins CD32B and CD79BClinical trials:
SLE
[31]
mAb = monoclonal antibody; RA = rheumatoid arthritis; GPA = Granulomatosis with Polyangitis; MPA = Microscopic Polyangitis; PV = Pemphigus Vulgaris; ITP = idiopathic thrombocytopenic purpura; MG = myasthenia gravis; GCA = giant cell arteritis; SSc-ILD = systemic sclerosis-interstitial lung disease; PJIA = polyarticular juvenile idiopathic arthritis (PJIA); SJIA = systemic juvenile idiopathic arthritis; MS = multiple sclerosis; CIS = clinically isolated syndrome; RRMS = relapsing–remitting MS; PPMS = primary progressive MS; SPMS = secondary progressive MS; IgG4-RD = IgG4-related diseases.
Table 2. CHO variant cell lines for afucosylated mAb productions.
Table 2. CHO variant cell lines for afucosylated mAb productions.
Cell LineAffected Biosynthesis PathwayReference
CHO Lec13 (Pro-Lec13.6a)Natural deficiency in endogenous GDP-mannose 4,6-dehydratase (GMD)[51,56]
CHO-DG44 FUT8−/− (BioWa)FUT8 knockout by homologous recombination[57]
Patent# US6946292B2
CHO-K1 FUT8−/−FUT8 deletion by ZFN[58]
CHO-gmt3 (CHO-glycosylation mutant3)GDP-fucose transporter (SLC35C1) inactivation[59]
CHO-RMDHeterologous expression of GDP-6 deoxy-d-lyxo-4-hexulose reductase (RMD) in the cytosol of CHO cells to deflect the GDP-fucose de novo pathway[60]
CHO-GnT-IIIOverexpressed GnTIII
catalyzes the formation of a bisecting GlcNAc to reduce Fc core fucosylation
[61]
CHO-SMGDP-fucose 4,6-dehydratase (GMD) knockout, which makes the cell unable to produce intracellular GDP-fucose and fucosylated glycoproteins in the absence of L-fucose[62]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Qian, Y.; Mahmoud, T.I. Molecule Formats of B-Cell Targeting Biologics: Applications in Autoimmune Disease Treatment and Impacts on Manufacturability. Pharmaceutics 2025, 17, 495. https://doi.org/10.3390/pharmaceutics17040495

AMA Style

Qian Y, Mahmoud TI. Molecule Formats of B-Cell Targeting Biologics: Applications in Autoimmune Disease Treatment and Impacts on Manufacturability. Pharmaceutics. 2025; 17(4):495. https://doi.org/10.3390/pharmaceutics17040495

Chicago/Turabian Style

Qian, Yueming, and Tamer I. Mahmoud. 2025. "Molecule Formats of B-Cell Targeting Biologics: Applications in Autoimmune Disease Treatment and Impacts on Manufacturability" Pharmaceutics 17, no. 4: 495. https://doi.org/10.3390/pharmaceutics17040495

APA Style

Qian, Y., & Mahmoud, T. I. (2025). Molecule Formats of B-Cell Targeting Biologics: Applications in Autoimmune Disease Treatment and Impacts on Manufacturability. Pharmaceutics, 17(4), 495. https://doi.org/10.3390/pharmaceutics17040495

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop