You're using an outdated browser. Please upgrade to a modern browser for the best experience.
BCMA-Targeting Antibody-Drug Conjugates in Multiple Myeloma: Comparison
Please note this is a comparison between Version 1 by Yuntong Liu and Version 2 by Beatrix Zheng.

Multiple myeloma (MM) is an incurable cancer of the plasma cells. OvIn ther the last twenty years, treatment strategies have evolved toward targeting MM cells—from the shotgun chemotherapy approach to the slightly more targeted approach of disrupting important MM molecular pathways to the immunotherapy approach that specifically targets MM cells based on protein expression. Antibody-drug conjugates (ADCs) are introduced as immunotherapeutic drugs which utilize an antibody to deliver cytotoxic agents to cancer cells distinctively. Recent investigations of ADCs for MM treatment focus on targeting B cell maturation antigen (BCMA), which regulates B cell proliferation, survival, maturation, and differentiation into plasma cells (PCs). Given its selective expression in malignant PCs, BCMA is one of the most promising targets in MM immunotherapy. Compared to other BCMA-targeting immunotherapies, ADCs have several benefits, such as lower price, shorter production period, fewer infusions, less dependence on the patient’s immune system, and they are less likely to over-activate the immune system. 

  • B cell maturation antigen
  • multiple myeloma
  • antibody-drug conjugates
  • Anti-BCMA ADC
  • drug resistance

1. B Cell Maturation Antigen (BCMA)

B cell maturation antigen (BCMA) was first identified in 1992 on the short arm of chromosome 16 at 16p13.1 in malignant human T-cell lymphoma [1][28]. It is a type III transmembrane glycoprotein with 6 conserved cysteines in its extracellular domain. It belongs to the tumor necrosis factor receptor (TNFR) superfamily as TNRSF17 and is primarily present in a perinuclear structure that overlaps the Golgi apparatus, but functional BCMA is also found on the cell surface [1][2][3][4][28,29,30,31].
BCMA functions in conjunction with two related TNFR superfamily members, B-cell activation factor receptor (BAFF-R) and transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI). Their collaboration regulates various aspects of B cell activities, such as proliferation, survival, maturation, and differentiation into plasma cells (PCs) [2][3][5][29,30,32]. Upon binding its cognate ligands, BAFF and APRIL, BCMA can activate the NF-kB, Elk-1, p38, or JNK pathways to transduce signals for corresponding functions [4][6][7][8][31,33,34,35]. Conversely, a soluble form of BCMA (sBCMA), generated by γ-secretase (GS), neutralizes APRIL as a decoy and hinders the activation of subsequent BCMA pathways [9][36]. The sBCMA level has been suggested as a biomarker since it is significantly higher in MM patients compared to healthy individuals, and higher levels are associated with poor prognosis [10][11][37,38], MM progression, and poor response to BCMA-targeted therapy [11][12][38,39].
Since its discovery, various studies have demonstrated that BCMA is a promising immunotherapeutic target for multiple myeloma. BCMA expression is restrictively found on the surface of plasmablasts and differentiated PCs, with no expression on CD34+ hematopoietic stem cells, naive B cells, memory B cells, and other normal tissue cells [13][40]. Furthermore, it has a high expression on the surface of the MM cell and is necessary for the survival of long-lived bone marrow plasma cells [14][15][41,42]. Both BCMA mRNA and protein have higher expression in malignant PCs than normal PCs, as validated by multiple gene expression profiling [13][16][17][40,43,44] and immunohistochemistry studies [13][40]. Moreover, Carpenter et al. [13][40] found BCMA cDNA in several hematologic tissues, including blood leukocytes, bone marrow, spleen, lymph node, and tonsil, but no BCMA cDNA in other normal human tissues except for the testis, trachea, and some gastrointestinal organs, where low levels of BCMA cDNA were detected, likely from plasma cells present in lamina propria and Peyer’s patches. These findings indicate BCMA is a desirable therapeutic target.

2. Anti-BCMA ADC

An antibody-drug conjugate (ADC) solves many of the above problems. An ADC comprises three components: a monoclonal antibody against a target, a cytotoxic agent (payload), and a stable linker connecting the two [18][19][20][54,55,56]. Since only a small amount of injected antibodies localize to tumor cells, most payloads are highly potent, with cytotoxicity in the picomolar range, often targeting tubulin or causing DNA damage [20][21][22][56,57,58]. The linker covalently binds the payload to the antibody and is critical to ADC efficacy, pharmacokinetics, pharmacodynamics, and therapeutic index. A stable linker ensures the release of the cytotoxic drug to target tissue and minimizes toxic effects. On the other hand, an overly strong linker impedes the delivery of the drug. Both cleavable and non-cleavable linkers, which rely on the physiological environment and degradation in endosomes and lysosomes, respectively, have been developed [23][24][59,60].
In MM treatment, BCMA is considered one of the most promising targets of ADCs. Following binding to BCMA, the ADC is internalized by endocytosis. The drug is released by cleavage or degradation in endosomes or lysosomes and then causes DNA damage, inhibits transcription, or disrupts microtubules, which leads to apoptosis (Figure 1). The development of anti-BCMA ADCs is an active area of research, and several anti-BCMA ADCs are in various stages of clinical trials (Table 1).
Figure 1.
General mechanism action of an anti-BCMA ADC in myeloma cell. Created with BioRender.com.
Academic Video Service