Next Article in Journal
Solvent Performance Evaluation of Heavy Oil in Coal–Oil Co-Liquefaction
Previous Article in Journal
Effects of High Glucose on Simulated Ischemia/Reperfusion Injury in Isolated Cardiomyocytes
Previous Article in Special Issue
Distinctive Features of Extracellular Vesicles Present in the Gastric Juice of Patients with Gastric Cancer and Healthy Subjects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targets for CAR Therapy in Multiple Myeloma

by
Olga A. Bezborodova
*,
Galina V. Trunova
,
Elena R. Nemtsova
,
Varvara A. Khokhlova
,
Julia B. Venediktova
,
Natalia B. Morozova
,
Maria S. Vorontsova
,
Anna D. Plyutinskaya
,
Elena P. Zharova
,
Peter V. Shegai
and
Andrey D. Kaprin
Moscow Hertsen Research Institute of Oncology—Branch of the National Medical Radiology Research Center, Ministry of Health of the Russian Federation, Moscow 125284, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(13), 6051; https://doi.org/10.3390/ijms26136051
Submission received: 22 May 2025 / Revised: 19 June 2025 / Accepted: 19 June 2025 / Published: 24 June 2025
(This article belongs to the Special Issue Advancements in Cancer Biomarkers)

Abstract

Multiple myeloma (MM or plasma cell myeloma) is a heterogenous B-cell malignant tumor that typically exhibits a high recurrence rate, resistance to drugs, and molecular diversity of tumor subclones. Given the limited efficacy of standard therapy options, cellular immunotherapy featuring a chimeric antigen receptor (CAR) has proven tangible potential in treatment for relapsed and refractory forms of MM. The rational choice of a tumor target which shows high selectivity, stable expression, and biological significance is key to the successful implementation of CAR therapy. This review has summarized and analyzed data from the literature on biological properties, the features of expression, and the clinical development stages of CAR cell products for MM treatment which target BCMA, GPRC5D, FcRH5, SLAMF7, CD38, CD138, TACI, APRIL, CD19, TNFR2, CD44v6, CD70, NKG2D ligands, etc. Special focus is on strategic approaches to overcoming antigenic escape, such as multi-specific CAR constructs, logical activation sequences, and controlled safety systems. The analysis underscores the need for integrating the molecular selection of targets with cutting-edge bioengineering solutions as a key trend for raising the efficacy, stability, and safety of cellular therapy in the case of MM.

Graphical Abstract

1. Introduction

Multiple myeloma (MM) is the second most widespread blood cancer disease and exhibits pronounced molecular heterogeneity, aggressive clinical progression, and a high risk of drug resistance occurrence [1,2].
Contemporary MM treatment regimens include the use of target drugs, such as primarily proteasome inhibitors, which have proven their high efficacy across different stages of the disease [3,4,5]. Nevertheless, the clinical application of such agents is limited by the development of drug resistance, including cross-resistance to other anti-tumor agents [6].
The utilization of immunotherapy with monoclonal antibodies, such as daratumumab and elotuzumab that are embedded in contemporary MM treatment standards and have become an integral part of combined therapy regimens, has evolved into an extra therapeutic area [7]. When combined with other drugs, this contributes to higher efficacy and partial overcoming of resistance but fails to solve the problem, i.e., completely eradicate the disease [8].
CAR (Chimeric Antigen Receptor)-based cellular therapy stands out as a breakthrough and promising approach to the treatment of relapsed and refractory MM. CAR T-cell therapy has shown a high frequency of therapeutic responses in patients that have used up the opportunities offered by standard treatment methods, which corroborates the tangible potential of the technology [9,10].
The B-cell maturation antigen (BCMA), which is expressed essentially on the surface of malignant plasma cells, appears as the best-known target for CAR therapy in the case of MM. However, the long-term efficacy of BCMA-targeted therapy is limited with the development of antigen-negative relapses and the depletion of effector T cells, which emphasizes the need for putting in place improved strategies and novel approaches to CAR creation [11,12].
This review is aimed to summarize and analyze data from the literature on biological properties, the features of expression, and the clinical stages of CAR cell products for MM treatment (Figure 1).

2. Structural Organization and Biological Fundamentals of CAR Cell Products in Immunotherapy of Blood Cancer Diseases

2.1. Background for CAR Cell Products in Therapy of MM

CAR cell product-based immunotherapy holds a key position among contemporary strategies for the treatment of blood cancer diseases such as MM due to its high specificity, capability for the targeted destruction of tumor cells, and efficacy in patients with resistant forms of the disease [13,14,15]. Central to this therapy are the chimeric antigen receptors (CARs), artificially constructed molecules that instill in T cells the capability for antigen-specific recognition, activation, and proliferation regardless of the major histocompatibility complex (MHC) molecules [16]. This mechanism is pivotal in tumor evolution and immune escape, since malignant cells oftentimes lose the expression of MHC molecules, thereby avoiding being recognized with traditional T-cell receptors [17].

2.2. Structural Organization of CAR Cell Products and Their Interaction with Tumor Antigen

The CAR construct incorporates three functional segments: an extracellular antigen-binding domain, a transmembrane domain, and intracellular signaling domains [18]. As a rule, the typical extracellular domain is a single-chain variable fragment of the scFv antibody with high specificity for tumor antigens [19]. The transmembrane domain acts as an anchor and fixes the CAR on the T-cell surface, facilitating its stable expression and the origination of multimeric complexes [20]. Intracellular signaling domains ensure the activation of a T cell in response to an interaction with the target, determining the range of its functional activity, including proliferation, the secretion of cytokines, and cytotoxicity [21].
The most well-known cellular products are used in contemporary cancer immunotherapy—CAR T cells and CAR NK cells, which differ by their cellular origin and constructional features of chimeric receptors [22,23,24].
The typical construct of CAR T cells uses CD3ζ—a component of the T-cell receptor (TCR) signaling pathway that ensures the imitation of the physiological activation of T lymphocytes as the intracellular signaling domain [25]. The co-stimulating domains CD28 and/or 4-1BB (CD137) are included into the CAR construct to enhance the antigen-specific response. CD28 ensures powerful primary activation of T cells and facilitates their quick expansion; in turn, 4-1BB maintains long-term survival and resistance to depletion and promotes the formation of memory T cells [16,26].
Typical constructs of CAR NK cells include signaling modules that are inherent for an innate immune system link—DAP10 (DNAX-activating protein 10) and DAP12 (DNAX-activating protein 12), as well as the co-stimulating receptor 2B4 (CD244). These elements initiate endogenous signaling cascades that are specific for the activation of NK cells, thus ensuring the effective cytotoxic activity and secretion of pro-inflammatory cytokines [27,28]. Unlike CAR T cells, CAR NK cells have a more controllable activation profile, which mitigates the risk origination of hyperactivation and a cytokine storm, thereby preserving pronounced anti-tumor efficacy.
In most CAR products, such as CAR T cells and CAR NK cells, a single-chain variable fragment of the scFv antibody is used as an extracellular antigen-binding domain [29]. However, some constructs of CAR NK cells use innate immunity receptors, for instance NKG2D (natural killer group 2D) and DNAM-1 (DNAX accessory molecule-1), as an extracellular domain. NKG2D recognizes NKG2D ligands (in total eight); among them two stress-associated molecules, MICA and MICB (MHC class I polypeptide-related sequence A/B), are expressed by tumor cells. The expression level of these proteins on tumor cells is higher than in untransformed ones, which makes them selective targets [30].
The tumor antigen primary recognition process with a chimeric receptor (CAR) starts with the interaction of the extracellular domain scFv with a specific epitope of a ligand molecule on the tumor cell surface [31]. The structural and chemical complementarity of scFv to the target epitope ensures high binding specificity, enabling it to effectively distinguish tumor cells and minimize damage to healthy cells. The following act as the key features in this interaction: affinity, i.e., the binding strength of one scFv with the epitope; avidity, i.e., the aggregate strength of multiple interactions between the CAR and antigens; and the uniqueness of a paratope, i.e., the individual properties of the antigen-binding center of a scFv. Together, these parameters play a crucial role in the functional outcome of the interaction between the effector and tumor cells, determining the strength, duration, and efficacy of the immune response [32].

2.3. Sources for Producing CAR Cell Products

Off-the-shelf CAR T cells are predominantly produced from autologous or allogeneic (donor) T lymphocytes that undergo successive release, activation, gene modification (transduction) and expansion stages prior to infusion into a patient. In CAR NK therapy the range of sources for obtaining the cell material are sizably wider. Adult peripheral blood, umbilical cord blood, the NK-92 cell line, and induced pluripotent stem cells (iPSCs) capable of differentiation into NK cells can be used to obtain NK cells. A variety of sources paves a way towards the development of personalized and standardized CAR NK products, such as ready-to-use off-the-shelf medications, which drastically expands the clinical opportunities and enhances the scalability of the therapy [28,33,34,35].

2.4. Safety of CAR Cell Products

Despite the high clinical efficacy in treatment for blood cancer diseases, CAR T-cell therapy is oftentimes accompanied by the development of grave side effects—cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) [13,36].
Unlike CAR T cells, CAR NK cells show a more favorable safety profile, stipulated by more balanced natural mechanisms for regulating the cytotoxic activity of NK cells and their reduced capability for uncontrollable production of pro-inflammatory cytokines [27,30,37].
The state-of-the-art condition of CAR cell therapy is characterized by rapid growth and high clinical efficacy, which is facilitated by solid achievements in genetic engineering of chimeric antigen receptors. Most sweeping innovations are related to the structural optimization of CAR T cells and CAR NK cells to the choice of the most suitable antigen targets to improve the safety profile of the therapy while preserving the stable anti-tumor activity. Nonetheless, despite the successes, the selectivity and stability of CAR interactions with tumor targets is still the key restrictive factor, as it determines both the immediate clinical effect and its duration.
To that effect, further evolution of this field is impossible without comprehensive engineering and molecular solutions with a view to overcome the immune escape mechanisms, boosting the affinity of the interaction and functional resistance of CAR cells.
Optimizing the “CAR—tumor target” axis is evolving as the pivotal area for prospective research, with the potential to substantially boost the efficacy and reliability of cellular immunotherapy.

2.5. Bioengineering Platforms and State-of-the-Art Strategies for Multi-Specific CAR Therapy in Cancer Immunotherapy

The development of poly-specific constructs that can enable a single population of T cells to simultaneously recognize several key tumor antigens has emerged as a priority area in contemporary cell engineering in the field of CAR therapy [38,39,40]. Such a strategy notably mitigates the risk of antigen escape, i.e., one of the basic mechanisms for tumor resistance to immunotherapy [41,42]. Moreover, poly-specific CAR platforms unveil new clinical prospects, including the potential for implementing the concept of personalized and adaptive cellular therapy that targets the molecular profile of a tumor in a specific patient [43].
Modern biotechnology strategies of obtaining biospecificity in CAR therapy pursue the creation of constructs that can recognize a bunch of tumor targets at once. The development of tandem CARs (TanCARs) with two or more successively located scFv domains is one of such approaches. Such extracellular domain architecture enables tandem CARS to simultaneously recognize various tumor antigens to improve the receptor-to-ligand bond strength, accuracy, and stability of the therapeutic response. An alternative is ligand-based CARs, which rely on the use of natural ligands in the extracellular domains of the receptor. An example is APRIL-CAR, which uses APRIL (a proliferation-inducing ligand) and is capable of binding with several receptors expressed on tumor cells—namely, BCMA and TACI (transmembrane activator and CAML interactor). Such an approach enables APRIL-CAR to expand the range of tumor target recognition via multi-ligand interactions while preserving the physiological specificity [44].
The development of bicistronic vectors that code various CARs facilitates the expression of two robust receptors in one cell, which promotes more effective targeting of tumor cells with a heterogenous profile of antigen expression [45]. An alternative approach, i.e., co-transduction, in which one T cell is modified for the expression of two or more CARs, ensures expanded antigen coverage of targets and may boost the efficacy of therapy in tumors with combined expression of antigens [46,47].
CAR constructs with logical activation elements, such as AND, NOT, and OR gates, have been developed in recent years to enhance selectivity and mitigate the risk of off-tumor toxicity. The AND gate concept states that CAR T-cell activation only occurs during the simultaneous recognition of two tumor antigens: one receptor initiates the activation signal, while the other implements co-stimulation [48]. NOT gates, which are implemented with the aid of inhibiting CARs (iCARs), function under the principle of suppressing the effector activity of a T cell during its interaction with antigens expressing on healthy cells and thereby ensure an extra safety control level. DiCAR constructs are made of several signaling domains and reliably inhibit the uncontrollable activity of effector cells. OR-gated CAR T cells are used to solve the problem of tumor cell heterogeneity. These cells are able to target various antigens on different tumor cells to provide better killing coverage [49,50,51].
Overcoming the immunosuppressive tumor microenvironment (TME) is a key problem for preserving the functional activity of CAR T cells in vivo [52]. For this purpose, armored CAR T cells of the third and subsequent generations are being developed; these have the capability to express the pro-inflammatory cytokines IL-12, IL-15, and IL-18 that augment the anti-tumor immune response. Such cells can additionally be modified for the expression of the dominant-negative transforming growth factor beta receptor II (TGF-βRII). TGF-βRII, which is expressed in CAR T cells, blocks the suppressive impact of TGF-β on T lymphocytes [53,54,55,56].
The next stage in the development of cellular therapy technologies is the creation of a fourth-generation CAR—the TRUCK platform (T cells redirected for universal cytokine killing), which ensures the local secretion of pro-inflammatory cytokines in response to a CAR binding with the tumor antigen [57,58]. This approach promotes the augmentation of the anti-tumor immune response and precludes the formation of an immunosuppressive TME [59,60,61].
Fifth-generation CAR T cells are additionally provided with intracellular domains that can activate signaling pathways associated with pro-inflammatory cytokines. These are elements of the JAK-STAT (Janus kinase—signal transducer and activator of transcription) signaling cascade, which ensures stable and autonomous cell activation even under unfavorable conditions [62].
Various molecular safety ‘switches’ that enable us to control the activity of modified T cells are integrated into the CAR construct to enhance the safety and controllability of the cellular therapy [63]. For instance, hypoxically inducible CARs activated solely under reduced oxygen conditions, typical for the tumor tissue, have been created. This approach ensures localized activation of CAR T cells and mitigates the risk of damage to normal tissues [64,65].
Relapses of the malignant process remain a frequent even after the minimal residual disease (MRD) state is achieved. As a rule, their occurrence is related to immune escape mechanisms due to the loss of target antigen expression by tumor cells and trogocytosis—the process in which the antigen is transferred to the CAR T-cell membrane and becomes unreachable for further recognition [66,67]. Multi-antigen CAR constructs capable of simultaneously recognizing several tumor targets, which drastically mitigates the risk of relapse occurrence and increases the stable effect of therapy, are being developed to overcome such restrictions [43,68].
The protocols based on combined or successful infusions of CAR products with varying antigen specificity levels attain extra clinical significance [69]. The use of similar treatment regimens is particularly promising in patients with refractory and relapsing forms of lymphomas, in whom successful therapy targeted at an expanded range of antigens promotes an increased frequency of remissions [70]. This novel therapeutic approach reduces the likelihood of relapse by eliminating the tumor clones with varying antigen expression levels, thereby minimizing the likelihood of immune escape [71,72].
In spite of the remarkable progress in the engineering of CAR constructs, the rational choice of a tumor target remains key to clinical efficacy. This problem becomes particularly relevant in pronounced molecular and phenotypic heterogeneity that is inherent for MM. In such cases, the stability of antigen expression on tumor cells, as well as their absence on normal tissues, has a direct impact on both the efficacy and safety of the therapy.

3. Tumor Targets in CAR Therapy for Multiple Myeloma

The rational choice of a tumor target is among the crucial stages in the development and clinical implementation of CAR cell therapy. It pursues the maximal therapeutic effect while simultaneously mitigating the risk of unwanted immunological complications, such as systemic toxicity and autoimmune reactions [73].
The presence of a tumor antigen that is capable of ensuring highly specific and reproducible recognition of pathological cells with minimal or absent expression in healthy tissues is key to the successful application of CAR therapy. This approach is critical to prevent the development of on-target, off-tumor toxicity—a hazardous phenomenon, in which CAR cells attack the body’s normal cells that express a low level of the target antigen. This phenomenon remains one of the main restrictions for the application of immunotherapy in the case of malignant neoplasms, including MM.
MM is characterized by pronounced molecular and phenotypic heterogeneity, which suggests the availability of multiple subclonal populations of tumor cells that differ by the level and range of surface antigen expression. This factor substantially complicates the choice of the single universal therapeutic target that can ensure an effective and selective impact [42,74].
When choosing an optimal antigen in the contemporary stage of CAR cell technology development, apart from its selectivity and expression level, several factors also account for a bunch of other critical parameters, including the biological role of the antigen in tumor pathogenesis and progression, expression stability in all subpopulations of myeloma cells, a link to the development of drug resistance, and the likelihood of cross-reactivity with normal tissues [75]. Moreover, the translational potential of a chosen target should also be analyzed for the scalability of manufacturing processes and clinical applicability, including the standardization and commercialization capability [76,77].
A comprehensive examination of tumor target features, including an analysis of their biological function, expression level, and stability on myeloma cells and an evaluation of their biological significance, clinical potential, and possible application restrictions as part of the CAR cell therapy, is a field in the development of more effective and safer MM treatment strategies [78,79]. Particularly noteworthy in this connection is reviewing the most promising targets with high utilization potential in the construct of CARs in the case of MM (Table 1).

3.1. B-Cell Maturation Antigen (BCMA)

BCMA, also known as TNFRSF17 (tumor necrosis factor receptor superfamily member 17), is a single-chain transmembrane protein that belongs to the tumor necrosis factor receptor superfamily (TNFRSF). Its construct includes an extracellular cysteine-rich domain (CRD), a transmembrane segment, and a short cytoplasmic domain [95,96]. These features of the construct stipulate for BCMA’s high selectivity as a therapeutic target for CAR cell therapy of MM, since the extracellular CRD ensures stable and specific binding with the scFv fragment of a CAR [97,98]. Standing as an extra benefit is the absence of tangible homology of BCMA’s extracellular area with other receptors, which mitigates the risk of cross-reactivity and on-target, off-tumor toxicity [99,100].
BCMA is expressed stably and with high density on the surface of malignant plasma cells, whereby it is virtually absent in other tissues, save for normal plasma cells [101]. This makes BCMA an attractive target not only for CAR cell therapy but also for other immunotherapy strategies.
At the molecular level, BCMA is engaged in the survival regulation of plasma cells. The interaction with the APRIL or BAFF (B-cell activating factor) ligand activates the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), PI3K/AKT (phosphatidylinositol-3-kinase/protein kinase B), and MAPK/ERK (mitogen-activated protein kinase/extracellular signal-regulated kinase) signaling cascades, which promote the growth, survival, and resistance of myeloma cells to apoptosis [101,102].
Clinical data corroborate that BCMA’s high expression level correlates with the intensity of the anti-tumor response, the likelihood of achieving complete remission, and the duration of relapse-free and overall survival [96,103].
Clinical trials, including KarMMa (for idecabtagene vicleucel, ide-cel) and CARTITUDE-1 (for ciltacabtagene autoleucel, cilta-cel), have shown high efficacy of BCMA-specific CAR T cells in patients with relapsing and refractory MM [104,105]. Idecabtagene vicleucel (ide-cel, Abecma; Bristol Myers Squibb Company, Summit, NJ, USA) is the first registered BCMA-specific CAR T product. The KarMMa trial reached an objective response rate (ORR) of 73%, complete remission (CR) in 33% of patients, a median overall survival duration of 19.4 months, and survival without progression—8.8 months. Side effects included CRS in 84% of patients (grade > 3 in 5%) and ICANS in 18% (grade > 3 in 3%) [103].
Ciltacabtagene autoleucel (cilta-cel, Carvykti; Legend Biotech Corporation, Somerset, NJ, USA) is the second-generation product that contains two scFv domains that recognize various BCMA epitopes, which ensures high avidity and efficacy [106,107]. The CARTITUDE-1 trial reached an ORR of 97%, and strict complete remission was observed in 67% of patients, whereas the median response duration exceeded 24 months. CRS emerged in 95% of patients (grade ≥ 3 in 5%) and ICANS in 21% of them, with mainly low or average severity.
Despite the impressive clinical results, the long-term efficacy of BCMA-specific CAR T therapy remains limited. The development of antigen-negative relapses resulting from the selective elimination of BCMA-positive cells and the subsequent survival of clones that lost their expression of this antigen is a key factor that precludes its stability [67]. The occurrence of disease relapses is linked to the existence of the soluble BCMA form (sBCMA) that circulates in the blood and competes with the membrane-bound BCMA for the CAR, thereby decreasing the efficacy of therapy and potentiating its immune escape [108,109].
To mitigate the risk of therapeutic inefficacy caused by antigen escape, poly-specific CAR products that simultaneously target the BCMA and extra tumor antigens, such as GPRC5D, FcRH5, and others, are being developed. Similar platforms expand the range of target antigens and boost the stability of the clinical effect due to more effective targeting of tumor cell heterogenous clones [97,99]. The development of high-affinity scFv domains is in the pipeline. These can selectively recognize solely the membrane-bound form of BCMA (mBCMA), excluding the interaction with its soluble form (sBCMA). This increases the specificity and efficacy of CAR cell activation [98,100].
Combined therapeutic strategies that stipulate the co-application of BCMA-targeted CAR T cells with immune checkpoint inhibitors, such as PD-1, and γ-secretase inhibitors, have been embedded in clinical practice. The latter promote a decrease in the level of soluble BCMA (sBCMA) and higher expression of membrane BCMA (mBCMA) on the surface of tumor cells, thereby boosting the efficacy of CAR T therapy [110,111].
Another novel field of development is the creation of armored CAR cells, which are stable in an immunosuppressive TME and capable of secreting pro-inflammatory cytokines, enhancing their proliferation and functional activity [78,112].
Along with autologous platforms, also progressing has been the field of creating allogenous off-the-shelf CAR T cells or CAR NK cells that target BCMA and are produced from iPSCs or from donor materials. These technologies enable us to standardize production, increase the affordability of therapy, and reduce its cost while ensuring controllable engineering modification to prevent allogenous immune reactions [112,113].
Hence, BCMA is worthily acting as the centerpiece among cellular therapy targets in MM due to a fusion of high specificity, stable expression, and its notable functional role in disease pathogenesis. The variety of approaches for the modification of BCMA-targeted platforms corroborates its strategic significance in existing promising therapeutic strategies.

3.2. G Protein-Coupled Receptor Class C Group 5 Member D (GPRC5D)

GPRC5D is a transmembrane protein falling within the superfamily of G protein-coupled receptors (GPCRs). It possesses a compact extracellular N-end domain, which makes it an easy target for coupling with the scFv contained in CARs [105,114]. GPRC5D’s high expression stability in myeloma cells and its accessibility on the tumor surface facilitate quick and effective interactions with CAR cells, ensuring reliable activation of the effector response [115,116].
GPRC5D expression outside the tumor tissue is restricted and mostly displayed increased expression in the stratified flat squamous epithelium, which mitigates the risk of on-target, off-tumor toxicity development [116,117].
The clinical and preclinical trials corroborate the favorable safety profile of GPRC5D-specific CAR products. Dermal toxicity related to GPRC5D expression in the epithelium is primarily registered in the mild and reversible form [118]. CRS is observed in 75–80% of patients, is mainly of severity grade I–II, and is well controlled [119,120]. ICANS is registered in less than 10% of patients and primarily flows in the mild form [117,121]. Hence, the toxicity profile of GPRC5D-targeted CAR products is deemed more favorable than in some BCMA-specific CAR systems [122].
For the first time, clinical efficacy was confirmed in the OriCAR-017 (phase 1, POLARIS) and CARTITUDE-2 trials, where the patients with relapsing or refractory MM, who had earlier received BCMA-targeted therapy, reached an overall response rate (ORR) of 70–90% [120,121].
The prevention of antigen escape is still a key problem in treatment for MM characterized by high heterogeneity. Bi-specific CAR products that are simultaneously targeting GPRC5D and BCMA, both in CAR-T and in the CAR-NK format, have been developed in this connection. Similar platforms display improved clinical outcomes and resistance to incidences of relapses [123,124]. Moreover, poly-specific constructs with the inclusion of IL-15 (armored CAR) and allogenous off-the-shelf CAR NK products have been actively promoted, which ensures additional affordability of the therapy, cost reduction, and potential for wide clinical application [125,126].
Hence, GPRC5D holds a prominent place among promising targets for cellular therapy of multiple myeloma, namely in patients with resistivity or relapsing after BCMA-specific therapy. The ongoing development of GPRC5D-targeted bi-specific and allogenous platforms opens up new opportunities for effective and safe treatment for this patient group.

3.3. Fc Receptor-Homolog 5 (FcRH5, CD307)

FcRH5 is a transmembrane glycoprotein that falls within the family of FcR-like receptors. It is mostly expressed on mature B cells, plasma cells, and malignant transformed cells in MM [111,127]. FcRH5’s restricted expression in normal conditions and its high abundance on tumor plasma cells substantiate its potential as a cellular therapy target, particularly in patients with a relapsing or refractory course of disease [128].
The extracellular segment of FcRH5 includes nine Ig-like domains, which represents a lengthy structure with multiple potential epitopes for coupling with CARs. Such architecture promotes high specificity and mitigates the risk of cross-reactivity [128]. The transmembrane domain of FcRH5 ensures solid fixation in the membrane and maintains stable expression on the surface of tumor cells—a critical condition for the stable effect of CAR therapy.
FcRH5 expression has been discovered in all subclones of myeloma cells, including those that originate after the loss of BCMA expression or resistance to BCMA-specific therapy [129]. This enables us to use FcRH5 as a target for second-line therapy and as a component for poly- and bi-specific platforms [130]. A series of preclinical trials has indicated that FcRH5-specific CAR T cells possess high cytotoxicity in in vitro and in vivo models, which results in the complete eradication of tumor cells without substantial toxic effects [111,128].
Also, FcRH5-targeted CAR NK platforms have been actively developed, and in preclinical models they demonstrate high specificity, a low alloreactivity risk, and a potential for large-scale off-the-shelf production [131]. The clinical trials of FcRH5-specific CAR T and FcRH5-targeted bi-specific antibodies show predictable and controllable toxicity profiles. The major side effects include CRS and ICANS, which are generally reversible and of moderate severity [132,133].
Due to its high expression and antigen stability, FcRH5 has been increasingly included into poly- and bi-specific CAR platforms (for instance, FcRH5+BCMA and FcRH5+GPRC5D), which is particularly promising for combating antigen escape in the case of tumor heterogeneity [39,134].
As indicated by preclinical trials, the inclusion of the co-stimulating domain 4-1BB (CD137) or ICOS (inducible t-cell costimulator) into FcRH5-targeted CAR constructs substantially improves the proliferative activity, persistence, and functional stability of effector cells in vivo. This ensures a steady and prolonged clinical effect, which is particularly crucial in refractory forms of MM [135,136,137,138].
In phase I–II clinical trials, FcRH5-specific CAR T cells and bi-specific antibodies have demonstrated a high overall response rate even in patients who had earlier been resistant to BCMA-targeted drugs [129]. Mostly moderate side effects have been revealed, which confirms the favorable profile of manageable toxicity.
Thus, FcRH5 has been viewed as the most promising and clinically meaningful target in the arsenal of cellular therapy for multiple myeloma. Its application as a target for new generations of CAR products enables us to expand the range of therapy, enhance resistance to relapsing, and provide treatment for patients who do not respond to BCMA-targeted strategies.

3.4. CD38 as a Therapeutic Target for Cellular Immunotherapy

CD38 is a type II transmembrane glycoprotein with a wide array of biological functions. It is engaged in intercellular adhesion and acts as a component in cellular signaling systems [139]. Owing to its enzymatic activity, CD38 is engaged in the regulation of metabolism, energy metabolism, and the innate and adaptive immune responses [96,140].
The molecular structure of CD38 includes a short cytoplasmic N-end fragment, a transmembrane domain, and a lengthy extracellular segment that is responsible for the catalysis of enzymatic reactions and the interaction with the extracellular matrix components [141].
CD38 has high and stable expression on the surface of myeloma plasma cells in all the disease stages, which makes it an attractive target for targeted therapy [142,143]. This marker is also expressed in a number of immune system normal cells (activated T and B lymphocytes, NK-cells, and monocytes) and in epithelial cells (for instance, type II alveolocytes) and lymphoid organs, which boosts the risk for the incidence of on-target, off-tumor toxicity.
CAR constructs that are able to recognize cells with high expression of CD38 typical for myeloma have been developed with a view to increase selectivity and decrease toxicity [65,144]. Likewise constructs with controllable inactivation of CAR cells (safety switches), have been developed—for instance, inducible apoptotic cascades (iCasp9) or pharmacologically controlled suppression of CAR signaling [145,146].
Several preclinical trials have displayed the high cytotoxic activity of CD38-specific CAR T cells both for in vitro and in vivo models [146]. Results of phase I clinical trials (for example, NCT03473496) have confirmed the relative safety and efficacy of CD38-specific CAR T cells in patients with relapsing and refractory multiple myeloma [96,147,148].
The creation of bi-specific CAR constructs, which can simultaneously recognize CD38 and BCMA, is a promising field. Similar platforms show a synergistic effect, boosting efficacy and decreasing the likelihood of resistance occurrence [149,150].
Altogether, the further optimization of CARs towards higher efficacy and safety and the adoption of poly-specific approaches unveil new opportunities for expanding the therapeutic window and clinical applicability of CD38-specific products in multiple myeloma.

3.5. Signaling Lymphocytic Activation Molecule Family 7 (SLAMF7)

SLAMF7/CD319 is a transmembrane glycoprotein from the family of SLAM receptors, playing a crucial role in the coordination of the innate and adaptive immune response [151]. The expression of SLAMF7 has been mainly observed on plasma cells, NK cells, dendritic cells, and individual subpopulations of T lymphocytes while nearly absent on hematopoietic stem cells, which makes the molecule an attractive target for cellular immunotherapy [152].
The SLAMF7 construct includes two extracellular immunoglobulin-like domains—a distal V domain and a proximal C2 domain, both engaged in intermolecular interactions and recognition by effector cells [153,154]. As indicated by preclinical trials, both domains may serve as independent epitopes for the creation of CAR constructs with varying functional profiles. However, the high expression of SLAMF7 on NK cells creates an autocytotoxicity risk from SLAMF7-targeted CAR T cells, which may weaken the natural immune surveillance and increase the likelihood of infectious complications [9,155,156].
To minimize the risk, various modifications of both CAR T and CAR NK platforms have been implemented using genome editing technologies and the introduction of inhibiting signaling domains that enable the preservation of the anti-tumor activity [157].
The efficacy and high specificity of SLAMF7-specific CAR T cells has been corroborated by both preclinical models and in early phases of clinical trials. In a phase I trial (NCT03710421), the overall response rate (ORR) exceeded 50%, including cases of complete and strict complete remission. CRS was mainly observed in the mild or moderate form, without multiple episodes of heavy neurotoxicity [147,158,159].
Alongside CAR-T, SLAMF7-targeted CAR NK-cells, which represent a promising platform due to reduced toxicity and the capability of delivering off-the-shelf products, have been actively developed.
Hence, the SLAMF7 glycoprotein is a highly specific, biologically substantiated, and clinically proven target for cellular therapy in multiple myeloma. SLAMF7 inclusion into poly-specific CAR constructs that can simultaneously recognize several antigens may drastically boost anti-tumor efficacy and mitigate the risk of antigen escape.

3.6. CD138 (Syndecan-1) as a Therapeutic Target for Cellular Immunotherapy

CD138 (syndecan-1) is a transmembrane heparan sulfate proteoglycan falling within the syndecan family. It plays a key role in the regulation of cellular adhesion and migration, transferring signals inside the cell in coupling with growth factors, morphogens, chemokines, enzymes, and extracellular matrix components [160].
The extracellular domain of CD138 contains heparan and chondroitin sulfate chains that ensure an interaction with extracellular matrix components. The transmembrane component fixes a molecule in the membrane, whereas a highly restricted short cytoplasmic section is engaged in signal transfer via the cytoskeleton. Such a structure enables the engagement of CD138 in a wide range of physiological and pathological processes, including tumor growth and invasion. The high accessibility of the extracellular section makes CD138 an easy target for coupling with CARs and the subsequent activation of T-cell effector functions [151].
CD138 is distinct for its stable and intense expression on MM cells [161,162]. Normally, protein is expressed on mature plasma and epithelial cells, which is used in diagnostics, whereas its expression on hematopoietic stem cells is minimal, which mitigates the risk of damage to the hematological system’s regenerative pool [163,164]. However, CD138 presence on the epithelium of various organs may stimulate the growth of off-target toxicity when using CD138-specific CAR T-cells.
Engineering strategies are being adopted to minimize such effects: lower affinity CARs are used, as they are capable of differentiating the tumor and normal cells by antigen expression level [144], as well as systems for the controllable shutdown of CAR cells (safety switches) in the development of undesired toxicity.
Preclinical trials have identified high-cytotoxicity Cd138-targeted CAR T cells, both in vitro and in vivo [161]. Clinical data, including phase I testing (NCT03778346), have confirmed the acceptable safety profile: As a rule, CRS was mild; neurotoxicity was rare, and the response level varied from partial to complete and strict complete remissions [164]. Moderate persistence of CAR cells was observed, which emphasizes the need for the optimization of their viability and functional robustness [162].
Thus, CD138 remains a meaningful target for cellular therapy in multiple myeloma. Despite potential restrictions, further evolution of CAR engineering and streamlining of effector cell activation control open up the prospects for enhancing the efficacy and clinical applicability of CD138-specific approaches.

3.7. CD70 as a Therapeutic Target for Cellular Immunotherapy

CD70 is a transmembrane protein from the TNF receptor family. It is expressed primarily on activated B and T lymphocytes, on dendritic cells, and on a series of malignant transformed cells, including plasma cells in MM [165].
The unique molecular characteristics of CD70 stipulate for its high selectivity as a target for cellular therapy. Namely, CAR constructs that are based on the CD70 interaction with its natural ligand CD27 demonstrate high rates of expansion and persistence of effector cells in vivo, which are required to maintain the steady anti-tumor effect [166].
Preclinical trials have confirmed the high cytotoxic activity of CD70-specific CAR T cells with respect to myeloma cells, whereby the low toxicity of normal tissues has been noted, including hematopoietic stem cells, thanks to minimal CD70 expression of extratumor or activated immune cells [167,168]. Further optimization of CAR constructs via molecular modeling of the CD70-CD27 interaction has enabled us to enhance their proliferation, survival, and specificity in the action of modified T cells.
Allogenous CD70-specific CAR T products that are constructed using CRISPR/Cas9 technology have been actively developed in recent years. They pass clinical evaluations in patients with refractory MM forms and solid tumors (CTX131 and NCT04554425 trials), demonstrating the acceptable safety profile in early phases [169,170].
The development of CD70-targeted CAR NK cells has been underway. Preclinical trials have demonstrated their high specificity and efficacy. The first clinical data corroborate the potential of this approach for MM therapy, especially in the case of CD19-negative forms of the disease [171,172,173].
Dual-targeting strategies that are simultaneously aimed at CD70 and BCMA and aid to overcome clonal heterogeneity and minimize the risk of antigen escape have been particularly interesting [174].
Hence, CD70 is a highly specific and biologically substantiated target for cellular therapy in multiple myeloma. The integration of CD70-targeted CAR T and CAR NK platforms, including bi-specific constructs, opens up prospects for increasing therapeutic efficacy, deepening and prolonging remission in patients with relapsing and refractory MM forms.

3.8. NKG2D Ligands (NKG2DLs) as a Target for Cellular Therapy of Multiple Myeloma

NKG2D ligands are stressed-induced molecules that include MICA, MICB, and the ULBP protein family (UL16-binding proteins) [175]. These molecules are expressed on the surface of cells in response to stresses, such as DNA damage, hypoxia, viral infection, and tumor transformation [176]. With respect to expression in normal tissues, the level of NKG2D ligands remains very low. Such selectivity makes the molecules promising targets for immunotherapy of multiple myeloma (MM).
The NKG2D receptor is expressed on NK cells, γδ-T cells, and CD8+ cytotoxic T lymphocytes, enabling the interaction between innate and adaptive immunity [177]. The binding of NKG2D ligands on the surface of tumor cells with the receptor on effector immune cells triggers powerful immune activation and destruction of tumor cells even in the case of phenotypic heterogeneity of the tumor [178]. This mechanism helps overcome antigen escape, attack various tumor clones, and maintain steady immune control.
NKG2D-based CARs enable the activation of innate immune recognition mechanisms. The constructs that utilize the full NKG2D receptor ensure a wide range of action versus phenotypically heterogenous myeloma cells [175,176]. In early-phase clinical trials, the efficacy of NKG2D-CAR T cells has been demonstrated in patients with acute myeloid leukemia, myelodysplastic syndrome, and relapsing/refractory MM. The application of full NKG2D receptor-based autologous CAR T cells with the CD3ζ signaling domain has indicated good tolerability and clinical activity signs [179].
The development of improved constructs has been underway, which include co-stimulating modules, such as DAP10 and DAP12. This is aimed at the enhancement of metabolic activity, resistance to depletion, and the prolonged persistence of CAR cells in the immunosuppressive tumor microenvironment [2,78].
Thus, the system of NKG2D-NKG2D ligands is a versatile and selective target for MM cellular therapy. It has been particularly relevant in pronounced tumor heterogeneity and the augmentation of therapeutic resistance, ensuring the potential for the development of steady and effective treatment strategies.

3.9. CD19 as a Therapeutic Target for Cellular Immunotherapy

CD19 is a transmembrane glycoprotein from the immunoglobulin superfamily and is expressed primarily on the membrane of mature and immature B lymphocytes [180]. It plays a key role in the activation, proliferation, and survival of B cells, functioning as a co-receptor in the composition of the B-cell receptor (BCR) signaling complex, which makes it a critical element for immune response regulation [181,182].
Despite the fact that MM is falling within the tumors of plasma cells, some surveys describe subpopulations of myeloma cells with residual expression of CD19, the presence of which is associated with an unfavorable forecast, relapses, and resistance to therapy [39,183,184]. This justifies the interest for CD19 as an extra therapeutic target within the scope of multi-target approaches.
The antigen escape phenomenon remains one of the main problems. It is related to mutations, expression disorder, and alternative splicing of mRNA or disorder in the formation of disulfide bridges, which results in the loss of CD19 expression [185,186]. Nonetheless, in preclinical trials CD19-specific CAR T cells have demonstrated pronounced activity versus CD19-positive subclones of myeloma cells, both in the mono-specific format and as parts of tandem constructs jointly with anti-BCMA CARs [187,188]. Moreover, such cell products have revealed selectivity versus the tumor cells with minimal impact on the normal subpopulations of immune cells [2].
Dual- (CD19 and BCMA) and poly-specific targeting platforms have been actively developed in recent years, which enables us to effectively overcome intraclonal heterogeneity, prevent antigen escape, and minimize the MRD growth rate [189,190,191,192]. Likewise the strategies that provide for successive or combined injection of CD19- and BCMA-specific CAR products have been emerging, especially to patients after deep tumor reduction in BCMA-targeted therapy, where resistant CD19-positive clones survive [193,194,195].
Based on the accumulated preclinical and clinical data, CD19 has been viewed as a promising target for application in multi-target cellular immunotherapy in MM patients, notably in the occurrence of relapses and resistance to BCMA-targeted treatment [138].

3.10. Transmembrane Activator and CAML Interactor (TACI, TNFRSF13B)

TACI is a transmembrane receptor from the tumor necrosis factor (TNF) receptor superfamily and plays a key role in the regulation of plasma cell survival, differentiation, and homeostasis. In physiological conditions, TACI expression is primarily observed on activated B lymphocytes and plasma cells, where it performs immunoregulatory functions via its interaction with BAFF (B-cell activating factor) and APRIL (a proliferation-inducing ligand) [196]. The binding of these ligands with TACI activates the intracellular signaling cascades that regulate the production of antibodies, maintaining the survival of long-lived plasma cells and B-cell homeostasis.
TACI is often co-expressed with BCMA on the surface of malignant plasma cells in MM. As indicated by the trials, the co-expression of these molecules is observed in most of the myeloma clones, whereby TACI expression may be higher and more stable in the later stages of the disease [129,138]. These features acquire special significance against the background of a renowned escape mechanisms via BCMA loss, since it makes TACI an attractive standby therapeutic target [197].
Surveys on TACI-specific CAR T cells confirm their high efficacy versus TACI-positive MM cells in in vitro and in vivo systems [198]. These cells demonstrate quick expansion, steady persistence, pronounced tumor eradication, and a possibility for disease control during heterogenous expression of targets [199]. Thus, toxicity for untransformed cells is revealed at the minimal level.
This is the reason for the active adoption of bi-specific CAR constructs that simultaneously target BCMA and TACI [197,200]. Such an approach minimizes the risk of relapse caused by antigen escape and ensures a wide coverage of tumor subclones. State-of-the-art developments include the creation of fully humane CAR constructs and armored CAR T cells with enhanced signaling activity that are capable of overcoming the immunosuppressive impact of the tumor environment.
Hence, TACI inclusion as a target for CAR T therapy in MM is a promising field, corroborated by molecular logic and clinical relevance. The combined targeting of BCMA and TACI opens up opportunities for enhancing the depth and duration of remissions, namely in patients with a high relapse risk and heterogenous antigen expression.

3.11. A Proliferation-Inducing Ligand (APRIL)

APRIL is a part of the TNF family and appears as a natural ligand for BCMA and TACI receptors [201]. The interaction of APRIL with these receptors is fundamental for maintaining the viability, differentiation, and homeostasis of plasma cells, which is critical for their long-term functioning [202].
Since APRIL shows high affinity to BCMA and TACI receptors, CAR constructs have been developed, in which the modified APRIL is used as an antigen-binding domain. This architecture is beneficial for the capability of simultaneously targeting two targets with no need to apply complex, in terms of engineering, bi-specific antibodies. This simplifies the structure of CAR platforms and increases their versatility and flexibility [203].
As indicated by preclinical trials, APRIL-CAR T cells effectively destroy tumor clones that express BCMA, TACI, or both the markers, which ensures a wider coverage of tumor heterogeneity [204]. Such dual targeting mitigates the risk of antigen escape—a key restriction for conventional CAR T platforms—and helps achieve deep and steady remission [205].
Additional optimizations for the spatial configuration of the APRIL domain in the CAR construct, as well as enhancing signaling modules (for instance, the inclusion of the co-stimulating CD28 or 4-1BB domain), have enabled a substantial increase in persistence and functional activity of effector cells, especially in the immunosuppressive microenvironment, which is typical for MM [206].
Hence, APRIL-targeted CAR platforms form one more promising field for cellular therapy of multiple myeloma, ensuring expansion of the range, a decrease in the relapse likelihood, and enhancement of the overall treatment efficacy.

3.12. Tumor Necrosis Factor Receptor 2 (TNFR2, TNFRSF1B)

TNFR2 is a transmembrane protein of the TNF receptor family. Unlike TNFR1, its expression is restricted within a few cell types, such as regulatory T cells (Tregs); myeloid-derived suppressor cells; the endothelium; tumor cells with diverse histogenesis; including MM cells; and microenvironment cells [207,208]. Such selective expression underpins the key role of TNFR2 in the formation of the immunosuppressive microenvironment and maintaining the functional activity of Tregs, which contribute to tumor resistance to immune surveillance [209].
The TNFR2 construct includes four extracellular cysteine-rich domains (CRDs), a transmembrane section, and an intracellular domain, and it lacks a death domain, which differentiates it from TNFR1. This enables the activation of alternative signaling cascades, including NF-κB and PI3K/AKT, which contribute to survival, proliferation, and immunoregulation [210]. The presence of several CRDs makes TNFR2 a promising target for high-affinity CAR constructs.
TNFR2-specific CAR T cells demonstrate the ability to suppress the growth of TNFR2-positive tumors via the remodeling of the tumor microenvironment and the suppression of Tregs [211,212]. Moreover, bi-specific CAR platforms that simultaneously recognize TNFR2 and other antigens, such as BCMA or GPRC5D, which boost selectivity and mitigate the risk of antigen escape, have been developed [213].
The creation of armored CAR products with resistance to immunosuppressive factors is aimed at the enhanced functional stability of cells and proliferative potential in unfavorable tumor environments [214]. The promising outlook of such an approach is confirmed by results from early-phase clinical trials, which evaluate the safety of TNFR2-CAR T in patients with relapsing tumors, including MM [215]. Moreover, TNFR2-specific CAR NK cells capable of effectively infiltrating the tumor and destroying the TNFR2-positive cells by secretion of interferon-γ and granzymes are undergoing active development [208,212].
Despite predominantly tumor-associated expression, TNFR2 is represented on the surface membrane of Tregs and is engaged in the regulation of their activity. This fact requires careful attention—excessive suppression of the subpopulation may lead to the occurrence of autoimmune diseases. Therefore, we recognize the use of platforms with regulated activation and safety switch systems that ensure the controllable action of CAR cells [214].
In general, TNFR2 is a promising cellular immunotherapy target that is focused on overcoming immunosuppression, enhancing the anti-tumor response, and expanding the opportunities for personalized treatment of resistant multiple myeloma forms.

3.13. CD44v6 as a Target for Cellular Therapy of Multiple Myeloma

CD44v6 is a variant isoform of the CD44 transmembrane glycoprotein, and it is derived from alternative splicing with the inclusion of the v6 exon into the molecule’s extracellular domain [216]. This modification drastically alters the spatial structure of the extracellular section, enhancing its interaction with hyaluronic acid, metalloproteases, VEGF (vascular endothelial growth factor), and HGF (hepatocyte growth factor), which promotes the invasiveness and survival of tumor cells [217,218].
CD44v6 overexpression is typical for aggressive MM forms and is associated with disease progression. CD44v6 expression is observed in 17% of patients in early MM stages and reaches 43% in the case of plasma cell leukemia [219], whereby it is often linked with 13q14 deletion—an unfavorable prognostic marker [220].
CD44v6 is deemed as a promising target for immunotherapy owing to its restricted expression in normal tissues and its high abundance on tumor cells. However, its presence on keratinocytes and the epithelium of mucous membranes creates the risk for the occurrence of on-target, off-tumor toxicity, which requires the development of safe engineering solutions [221].
This is the purpose for applying low-affinity CAR constructs that are capable of differentiating cells with high and low Cd44v6 expression, as well as safety switch systems for urgent inhibition of effector cells if complications develop [222]. Particularly interesting are CAR-NK platforms, which demonstrate innate anti-tumor activity and a reduced rate of CRS incidence as compared with CAR T [24,223].
Preclinical trials have confirmed the high efficacy of both CD44v6-CAR T, and CAR NK cells versus CD44v6-positive tumors, without pronounced damage to normal tissues [224,225]. The first clinical trials (phases I/IIa) of CD44v6-targeted CAR products in patients with refractory hematological solid tumors, including MM, have demonstrated controllable safety profiles and preliminary efficacy [226,227]. CD44v6 as a target is a part of poly- and bi-specific CAR platforms and is used to overcome antigen escape and intratumoral heterogeneity [228].
Hence, CD44v6 is a prominent target for cellular therapy of multiple myeloma with high therapeutic potential, provided that meticulous engineering optimization of safety is conducted.

3.14. Alternative Targets for CAR Cell Therapy in Multiple Myeloma

Further, concerning the best known targets for CAR therapy in MM, preclinical trials on cellular products that target alternative molecules possessing therapeutic potential have been conducted [129]. These include CD56, an activated form of integrin β7 [229], CD123, and the tumor-associated antigen Lewis-Y [230]. The choice of these targets is due to restricted expression in normal tissues and engagement in MM pathogenesis, which make them attractive for the development of selective CAR cell constructs [124,231].
CD56, also known as neural cell adhesion molecule (NCAM), is expressed in more than 70% of MM patients and plays a crucial role in the migration and adhesion of tumor cells in the bone marrow [232,233]. The absence of CD56 expression is associated with a more aggressive course of multiple myeloma and a trend for increasing the quantity of plasma cells in the peripheral blood [234]. In normal tissues CD56 expression is primarily restricted to NK cells, which required a comprehensive safety evaluation in the development of CD56-specific CAR cells. Nevertheless, preclinical data have shown tangible anti-tumor activity of CAR T-cells targeted against this antigen [163].
Integrin β7, especially in its activated form, has been viewed as a promising therapeutic target, since it is primarily expressed on MM cells [235]. The use of the MMG49 monoclonal antibody, which selectively recognizes the activated form of integrin β7, has enabled us to engineer CAR constructs with high antigen specificity and mitigated the risk of off-tumor toxicity [235]. Preclinical trials have demonstrated the capability of such CAR cells to selectively lyse multiple myeloma cells while not affecting healthy hematopoietic cells [122].
CD123 (α-subunit of the interleukin-3 receptor) is an antigen which is expressed on tumor stem cells and plasmacytoid dendritic cells, and it is engaged in the formation of the immunosuppressive microenvironment in multiple myeloma [236,237]. Despite being actively investigated as a therapeutic target in acute myeloid leukemia, CD123 potential in therapy for multiple myeloma remains understudied [238,239]. Restricted CD123 expression in normal tissues opens up the prospects for its use in immunotherapy, but additional surveys are needed in light of possible hematological toxicity.
Lewis-Y is a tumor-associated carbohydrate antigen, and it is expressed on the surface of cells in various malignant neoplasms, including MM [240]. In preclinical trials Lewis-Y-specific CAR T cells have demonstrated pronounced anti-tumor activity [240,241]. A phase I clinical trial has shown cases of temporary remission and disease stabilization for a period of up to 23 months [242]. Moreover, the infiltration and prolonged persistence of CAR T cells (up to 10 months) has been confirmed, which is deemed a crucial factor for the efficacy of the therapy [230].
Despite the encouraging preclinical and limited clinical results, the scope of data accumulated on alternative targets yields BCMA-targeted strategies. The matters of toxicity, antigen escape, and therapeutic response stability have remained unsolved [129]. In this connection, the development of multi-antigen CAR constructs and engineering platforms with controllable activation of effector cells (for instance, systems like the AND gate), as well as the adoption of safety systems for the quick and controllable suppression of cytotoxic activity should any undesired effects occur, has been very relevant [2,243,244].
Hence, CD56, integrin β7, CD123, and Lewis-Y appear as biologically substantiated targets for further development of CAR cellular therapy for multiple myeloma. Their integration into clinical practice will require additional surveys with a focus on the safety, specificity, and long-term efficacy of the therapeutic response [122].

4. Conclusions

MM is characterized by a high relapse incidence and the formation of multiple drug resistance mechanisms. No radical recovery from the disease has been obtained so far, which necessitates the introduction of game-changing therapeutic approaches.
CAR cell therapy is an area with the most breakthroughs in treatment for relapsing and refractory forms of MM. Its efficacy is defined by multiple factors, with the rational choice of tumor target being the key one. The presented review has analyzed data on the biological properties, level, and stability of the expression, selectivity, and clinical development stages of target molecules, such as BCMA, GPRC5D, FcRH5, SLAMF7, CD38, CD138, TACI, APRIL, CD19, CD44v6, TNFR2, CD70, NKG2D ligands, etc.
BCMA is the most verified and clinically proven target for CAR cell therapy in MM. However, despite the high frequency of a therapeutic response, the use of this target shows some restrictions, including the development of antigen-negative relapses and the presence of the soluble BCMA form (sBCMA), which decreases the efficacy of cellular therapy.
The most promising targets in the advanced stage of clinical development include GPRC5D and FcRH5, which demonstrate high specificity to myeloma cells, the favorable safety profile, and efficacy in patients who had earlier received BCMA-specific therapy. The development of bi-specific CAR products that simultaneously target BCMA and GPRC5D/FcRH5 has shown encouraging results in overcoming antigen escape and increasing the stability of a clinical response.
Second-line targets, such as SLAMF7, CD38, CD138, TACI, and APRIL, possess biological validity, high expression on myeloma cells, and proven therapeutic potential in preclinical and early clinical trials. Meanwhile, the use of some targets requires particular care due to the risk of on-target, off-tumor toxicity or involvement in the regulation of immune homeostasis. These include CD44v6, TNFR2, CD70, and CD19, which are also expressed on normal epithelial or immune cells. CAR logical activation strategies (AND/NOT gates), low-affinity receptors, and controllable inactivation systems (safety switches) are used to increase therapeutic effect selectivity and controllability with respect to these targets.
Alternative targets, such as CD56, the activated form of integrin β7, CD123, and Lewis-Y, are primarily in the preclinical validation stage.
The choice of a tumor target in multiple myeloma is determined by a set of parameters. The optimization of antigen targeting via multi-specific CAR constructs, logically programmed platforms, and controllable safety mechanisms is a key field in the development of more effective, steady, and safe forms of cellular therapy in MM.

Author Contributions

Concept, writing, and discussion, O.A.B.; editing and discussion, G.V.T. and E.R.N.; selection of the literature and work with references, V.A.K., J.B.V., N.B.M., M.S.V. and A.D.P.; project administration, E.P.Z. and P.V.S.; funding acquisition, A.D.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Health of the Russian Federation under the Research. Project with the registration number 125012100524-9.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lee, H.; Neri, P.; Bahlis, N.J. BCMA- or GPRC5D-Targeting Bispecific Antibodies in Multiple Myeloma: Efficacy, Safety, and Resistance Mechanisms. Blood 2024, 143, 1211–1217. [Google Scholar] [CrossRef]
  2. Manier, S.; Ingegnere, T.; Escure, G.; Prodhomme, C.; Nudel, M.; Mitra, M.; Facon, T. Current state and next-generation CAR-T cells in multiple myeloma. Blood Rev. 2022, 54, 100929. [Google Scholar] [CrossRef] [PubMed]
  3. Manasanch, E.E.; Orlowski, R.Z. Proteasome Inhibitors in Cancer Therapy. Nat. Rev. Clin. Oncol. 2017, 14, 417–433. [Google Scholar] [CrossRef] [PubMed]
  4. Lu, Q.; Yang, D.; Li, H.; Niu, T.; Tong, A. Multiple Myeloma: Signaling Pathways and Targeted Therapy. Mol. Biomed. 2024, 5, 18. [Google Scholar] [CrossRef]
  5. Scalzulli, E.; Grammatico, S.; Vozella, F. Proteasome Inhibitors for the Treatment of Multiple Myeloma. Expert Opin. Investig. Drugs 2018, 27, 199–213. [Google Scholar] [CrossRef]
  6. Pinto, V.; Bergantim, R.; Caires, H.R.; Seca, H. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers 2020, 12, 407. [Google Scholar] [CrossRef]
  7. Varga, C.; Maglio, M.; Ghobrial, I.M. Current Use of Monoclonal Antibodies in the Treatment of Multiple Myeloma. Br. J. Haematol. 2018, 181, 447–460. [Google Scholar] [CrossRef]
  8. Larocca, A.; Mina, R.; Gay, F.; Bringhen, S. Emerging Drugs and Combinations to Treat Multiple Myeloma. Oncotarget 2017, 8, 60656–60672. [Google Scholar] [CrossRef] [PubMed]
  9. Gagelmann, N.; Riecken, K.; Wolschke, C.; Berger, C. Development of CAR-T Cell Therapies for Multiple Myeloma. Leukemia 2020, 34, 2317–2332. [Google Scholar] [CrossRef]
  10. Rasche, L.; Hudecek, M.; Einsele, H. CAR T-Cell Therapy in Multiple Myeloma: Mission Accomplished? Blood 2024, 143, 305–316. [Google Scholar] [CrossRef]
  11. Lee, H.; Ahn, S.; Maity, R.; Leblay, N.; Ziccheddu, B.; Truger, M.; Chojnacka, M.; Cirrincione, A.; Durante, M.; Tilmont, R.; et al. Mechanisms of Antigen Escape from BCMA- or GPRC5D-Targeted Immunotherapies in Multiple Myeloma. Nat. Med. 2023, 29, 2135–2145. [Google Scholar] [CrossRef]
  12. Rade, M.; Grieb, N.; Weiss, R.; Sia, J.; Fischer, L.; Born, P.; Boldt, A.; Fricke, S.; Franz, P.; Scolnick, J.; et al. Single-Cell Multiomic Dissection of Response and Resistance to Chimeric Antigen Receptor T Cells Against BCMA in Relapsed Multiple Myeloma. Nat. Cancer 2024, 5, 567–579. [Google Scholar] [CrossRef]
  13. Parikh, R.H.; Lonial, S. Chimeric Antigen Receptor T-Cell Therapy in Multiple Myeloma: A Comprehensive Review of Current Data and Implications for Clinical Practice. CA Cancer J. Clin. 2023, 73, 275–285. [Google Scholar] [CrossRef]
  14. Dhakal, B.; Hari, P.N.; Usmani, S.Z. Chimeric Antigen Receptor T Cell Therapy in Multiple Myeloma: Promise and Challenges. Bone Marrow Transpl. 2021, 56, 379–390. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, Z.; Chen, C.; Wang, L.; Jia, Y.; Qin, Y. Chimeric Antigen Receptor T-Cell Therapy for Multiple Myeloma. Front. Immunol. 2022, 13, 1050522. [Google Scholar] [CrossRef] [PubMed]
  16. Minguet, S.; Maus, M.V.; Schamel, W.W. From TCR Fundamental Research to Innovative Chimeric Antigen Receptor Design. Nat. Rev. Immunol. 2024, 24, 212–224. [Google Scholar] [CrossRef] [PubMed]
  17. Keshavarz, A.; Salehi, A.; Khosravi, S.; Shariati, Y.; Nasrabadi, N.; Kahrizi, M.S.; Maghsoodi, S.; Mardi, A.; Azizi, R.; Jamali, S.; et al. Recent Findings on Chimeric Antigen Receptor (CAR)-Engineered Immune Cell Therapy in Solid Tumors and Hematological Malignancies. Stem Cell Res. Ther. 2022, 13, 482. [Google Scholar] [CrossRef]
  18. Fujiwara, K.; Kitaura, M.; Tsunei, A.; Kusabuka, H. Structure of the Signal Transduction Domain in Second-Generation CAR Regulates the Input Efficiency of CAR Signals. Int. J. Mol. Sci. 2021, 22, 2476. [Google Scholar] [CrossRef]
  19. Hanssens, H.; Meeus, F.; De Veirman, K. The Antigen-Binding Moiety in the Driver’s Seat of CARs. Med. Res. Rev. 2022, 42, 306–342. [Google Scholar] [CrossRef]
  20. Fujiwara, K.; Tsunei, A.; Kusabuka, H.; Ogaki, E. Hinge and Transmembrane Domains of Chimeric Antigen Receptor Regulate Receptor Expression and Signaling Threshold. Cells 2020, 9, 1182. [Google Scholar] [CrossRef]
  21. Wang, P.; Wang, Y.; Zhao, X.; Zheng, R.; Zhang, Y.; Meng, R.; Dong, H.; Liang, S.; He, X.; Song, Y.; et al. Chimeric Antigen Receptor with Novel Intracellular Modules Improves Antitumor Performance of T Cells. Signal Transduct. Target. Ther. 2025, 10, 20. [Google Scholar] [CrossRef] [PubMed]
  22. Peng, L.; Sferruzza, G.; Yang, L.; Zhou, L. CAR-T and CAR-NK as Cellular Cancer Immunotherapy for Solid Tumors. Cell. Mol. Immunol. 2024, 21, 1089–1108. [Google Scholar] [CrossRef] [PubMed]
  23. Soldierer, M.; Bister, A.; Haist, C.; Thivakaran, A. Genetic Engineering and Enrichment of Human NK Cells for CAR-Enhanced Immunotherapy of Hematological Malignancies. Front. Immunol. 2022, 13, 847008. [Google Scholar] [CrossRef]
  24. Lu, H.; Zhao, X.; Li, Z.; Hu, Y.; Wang, H. From CAR-T Cells to CAR-NK Cells: A Developing Immunotherapy Method for Hematological Malignancies. Front. Oncol. 2021, 11, 720501. [Google Scholar] [CrossRef]
  25. Ruppel, K.E.; Fricke, S.; Köhl, U.; Schmiedel, D. Taking Lessons from CAR-T Cells and Going Beyond: Tailoring Design and Signaling for CAR-NK Cells in Cancer Therapy. Front. Immunol. 2022, 13, 822298. [Google Scholar] [CrossRef]
  26. Zhang, H.; Zhao, P.; Huang, H. Engineering Better Chimeric Antigen Receptor T Cells. Exp. Hematol. Oncol. 2020, 9, 31. [Google Scholar] [CrossRef]
  27. Rafei, H.; Daher, M.; Rezvani, K. Chimeric Antigen Receptor (CAR) Natural Killer (NK)-Cell Therapy: Leveraging the Power of Innate Immunity. Br. J. Haematol. 2021, 193, 216–230. [Google Scholar] [CrossRef] [PubMed]
  28. Pan, K.; Farrukh, H.; Chittepu, V.C.S.R.; Xu, H.; Pan, C.X.; Zhu, Z. CAR race to cancer immunotherapy: From CAR T, CAR NK to CAR macrophage therapy. J. Exp. Clin. Cancer Res. 2022, 41, 119. [Google Scholar] [CrossRef]
  29. Ramírez-Chacón, A.; Betriu-Méndez, S.; Bartoló-Ibars, A.; González, A.; Martí, M.; Juan, M. Ligand-based CAR-T cell: Different strategies to drive T cells in future new treatments. Front. Immunol. 2022, 13, 932559. [Google Scholar] [CrossRef]
  30. Balkhi, S.; Zuccolotto, G.; Di Spirito, A.; Rosato, A. CAR-NK Cell Therapy: Promise and Challenges in Solid Tumors. Front. Immunol. 2025, 16, 1574742. [Google Scholar] [CrossRef]
  31. Martarelli, N.; Capurro, M.; Mansour, G. Artificial Intelligence-Powered Molecular Docking and Steered Molecular Dynamics for Accurate scFv Selection of Anti-CD30 Chimeric Antigen Receptors. Int. J. Mol. Sci. 2024, 25, 7231. [Google Scholar] [CrossRef] [PubMed]
  32. Mao, R.; Kong, W.; He, Y. The Affinity of Antigen-Binding Domain on the Antitumor Efficacy of CAR T Cells: Moderate Is Better. Front. Immunol. 2022, 13, 1032403. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Y.; Li, J.; Wang, Z.; Liu, Y.; Wang, T.; Zhang, M.; Xia, C.; Zhang, F.; Huang, D.; Zhang, L.; et al. Comparison of Seven CD19 CAR Designs in Engineering NK Cells for Enhancing Anti-Tumour Activity. Clin. Transl. Immunol. 2024, 13, e13683. [Google Scholar] [CrossRef]
  34. Wu, X.; Matosevic, S. Gene-Edited and CAR-NK Cells: Opportunities and Challenges with Engineering of NK Cells for Immunotherapy. Mol. Ther. Oncolytics 2024, 27, 224–238. [Google Scholar] [CrossRef]
  35. Guo, F.; Zhang, Y.; Cui, J. Manufacturing CAR-NK Against Tumors: Who Is the Ideal Supplier? Chin. J. Cancer Res. 2024, 36, 145–157. [Google Scholar] [CrossRef]
  36. Mansouri, V.; Yazdanpanah, N.; Rezaei, N. The Immunologic Aspects of Cytokine Release Syndrome and Graft Versus Host Disease Following CAR T Cell Therapy. Int. Rev. Immunol. 2022, 41, 649–668. [Google Scholar] [CrossRef]
  37. Jørgensen, L.V.; Christensen, E.B.; Barnkob, M.B. The Clinical Landscape of CAR NK Cells. Exp. Hematol. Oncol. 2025, 14, 63. [Google Scholar] [CrossRef] [PubMed]
  38. Li, W.; Zhang, B.; Cao, W.; Zhang, W.; Li, T.; Liu, L.; Xu, L.; Gao, F.; Wang, Y.; Wang, F.; et al. Identification of potential resistance mechanisms and therapeutic targets for the relapse of BCMA CAR-T therapy in relapsed/refractory multiple myeloma through single-cell sequencing. Exp. Hematol. Oncol. 2023, 12, 44. [Google Scholar] [CrossRef] [PubMed]
  39. Lakshman, A.; Kumar, S.K. Chimeric Antigen Receptor T-Cells, Bispecific Antibodies, and Antibody-Drug Conjugates for Multiple Myeloma: An Update. Am. J. Hematol. 2022, 97, 803–815. [Google Scholar] [CrossRef]
  40. Tan, M.S.Y.; Chen, Y.; Smith, E.L. Beyond BCMA: Newer Immune Targets in Myeloma. Blood Adv. 2024, 8, 4433–4445. [Google Scholar] [CrossRef]
  41. Schavgoulidze, A.; Cazaubiel, T.; Perrot, A. Multiple Myeloma: Heterogeneous in Every Way. Cancers 2021, 13, 1285. [Google Scholar] [CrossRef] [PubMed]
  42. Bartoszewska, E.; Tota, M.; Kisielewska, M.; Skowron, I. Overcoming Antigen Escape and T-Cell Exhaustion in CAR-T Therapy for Leukemia. Cells 2024, 13, 1596. [Google Scholar] [CrossRef] [PubMed]
  43. Han, X.; Wang, Y.; Wei, J.; Han, W. Multi-Antigen-Targeted Chimeric Antigen Receptor T Cells for Cancer Therapy. J. Hematol. Oncol. 2019, 12, 128. [Google Scholar] [CrossRef] [PubMed]
  44. Timmers, M.; Roex, G.; Wang, Y. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front. Immunol. 2019, 10, 1613. [Google Scholar] [CrossRef]
  45. Lam, N.; Finney, R.; Yang, S.; Choi, S.; Wu, X.; Cutmore, L.; Andrade, J.; Huang, L.; Amatya, C.; Cam, M.; et al. Development of a bicistronic anti-CD19/CD20 CAR construct including abrogation of unexpected nucleic acid sequence deletions. Mol. Ther. Oncolytics 2023, 30, 132–149. [Google Scholar] [CrossRef]
  46. Bachiller, M.; Barceló-Genestar, N.; Rodriguez-Garcia, A.; Alserawan, L.; Dobaño-López, C.; Giménez-Alejandre, M.; Castellsagué, J.; Colell, S.; Otero-Mateo, M.; Antoñana-Vildosola, A.; et al. ARI0003: Co-Transduced CD19/BCMA Dual-Targeting CAR-T Cells for the Treatment of Non-Hodgkin Lymphoma. Mol. Ther. 2023, 33, 317–335. [Google Scholar] [CrossRef]
  47. de Oliveira Canedo, G.; Roddie, C.; Amrolia, P.J. Dual-Targeting CAR T Cells for B-Cell Acute Lymphoblastic Leukemia and B-Cell Non-Hodgkin Lymphoma. Blood Adv. 2025, 9, 704–721. [Google Scholar] [CrossRef]
  48. Zhang, B.; Wu, J.; Jiang, H.; Zhou, M. Strategies to Overcome Antigen Heterogeneity in CAR-T Cell Therapy. Cells 2025, 14, 320. [Google Scholar] [CrossRef]
  49. Tousley, A.M.; Rotiroti, M.C.; Labanieh, L.; Rysavy, L.W. Co-Opting Signalling Molecules Enables Logic-Gated Control of CAR T Cells. Nature 2023, 617, 763–769. [Google Scholar] [CrossRef]
  50. Bangayan, N.J.; Wang, L.; Burton Sojo, G.; Noguchi, M.; Cheng, D.; Ta, L.; Gunn, D.; Mao, Z.; Liu, S.; Yin, Q.; et al. Dual-Inhibitory Domain iCARs Improve the Efficiency of the AND-NOT Gate CAR T Strategy. Proc. Natl. Acad. Sci. USA 2023, 120, e2312374120. [Google Scholar] [CrossRef]
  51. Ebert, L.M.; Yu, W.; Gargett, T.; Brown, M.P. Logic-Gated Approaches to Extend the Utility of Chimeric Antigen Receptor T-Cell Technology. Biochem. Soc. Trans. 2018, 46, 391–401. [Google Scholar] [CrossRef]
  52. Pievani, A.; Biondi, M.; Tettamanti, S. CARs Are Sharpening Their Weapons. J. Hematol. Oncol. 2024, 12, e008275. [Google Scholar] [CrossRef] [PubMed]
  53. Alabanza, L.M.; Xiong, Y.; Vu, B.; Webster, B.; Wu, D.; Hu, P.; Zhu, Z.; Dropulic, B.; Dash, P.; Schneider, D. Armored BCMA CAR T Cells Eliminate Multiple Myeloma and Are Resistant to the Suppressive Effects of TGF-β. Front. Immunol. 2022, 13, 832645. [Google Scholar] [CrossRef]
  54. Tran, T.M.; Thakuri, B.K.C. Armored TGFβRIIDN ROR1-CAR T Cells Reject Solid Tumors and Resist Suppression by Constitutively-Expressed and Treatment-Induced TGFβ1. J. Immunother. Cancer 2024, 12, e008261. [Google Scholar] [CrossRef] [PubMed]
  55. Brog, R.A.; Ferry, S.L.; Schiebout, C.T. Superkine IL-2 and IL-33 Armored CAR T Cells Reshape the Tumor Microenvironment and Reduce Growth of Multiple Solid Tumors. Cancer Immunol. Res. 2022, 10, 962–976. [Google Scholar] [CrossRef] [PubMed]
  56. Carcopino, C.; Erdogan, E.; Henrich, M.; Kobold, S. Armoring chimeric antigen receptor (CAR) T cells as micropharmacies for cancer therapy. Immunooncol Technol. 2024, 24, 100739. [Google Scholar] [CrossRef]
  57. Glienke, W.; Dragon, A.C.; Zimmermann, K.; Martyniszyn-Eiben, A.; Mertens, M.; Abken, H.; Rossig, C.; Altvater, B.; Aleksandrova, K.; Arseniev, L.; et al. GMP-Compliant Manufacturing of TRUCKs: CAR T Cells Targeting GD2 and Releasing Inducible IL-18. Front. Immunol. 2022, 13, 839783. [Google Scholar] [CrossRef]
  58. Chmielewski, M.; Hombach, A.A. Of CARs and TRUCKs: Chimeric Antigen Receptor (CAR) T Cells Engineered with an Inducible Cytokine to Modulate the Tumor Stroma. Immunol. Rev. 2014, 257, 19–34. [Google Scholar] [CrossRef]
  59. Thomas, S.; Abken, H. CAR T Cell Therapy Becomes CHIC: “Cytokine Help Intensified CAR” T Cells. Front. Immunol. 2023, 14, 1090959. [Google Scholar] [CrossRef]
  60. Silveira, C.R.F.; Corveloni, A.C.; Caruso, S.R. Cytokines as an Important Player in the Context of CAR-T Cell Therapy for Cancer: Their Role in Tumor Immunomodulation, Manufacture, and Clinical Implications. Front. Immunol. 2022, 13, 947648. [Google Scholar] [CrossRef]
  61. Chmielewski, M.; Abken, H. TRUCKs, the Fourth-Generation CAR T Cells: Current Developments and Clinical Translation. Adv. Cell Gene Ther. 2020, 3, 84. [Google Scholar] [CrossRef]
  62. Zheng, Z.; Li, S.; Liu, M.; Chen, C.; Zhang, L.; Zhou, D. Fine-Tuning Through Generations: Advances in Structure and Production of CAR-T Therapy. Cancers 2023, 15, 3476. [Google Scholar] [CrossRef] [PubMed]
  63. Zheng, Y.; Nandakumar, K.S.; Cheng, K. Optimization of CAR-T Cell-Based Therapies Using Small-Molecule-Based Safety Switches. J. Med. Chem. 2021, 64, 9577–9591. [Google Scholar] [CrossRef] [PubMed]
  64. He, H.; Liao, Q.; Zhao, C.; Zhu, C.; Feng, M.; Liu, Z.; Jiang, L.; Zhang, L.; Ding, X.; Yuan, M.; et al. Conditioned CAR-T Cells by Hypoxia-Inducible Transcription Amplification (HiTA) System Significantly Enhance Systemic Safety and Retain Antitumor Efficacy. J. Immunother. Cancer 2021, 9, e002755. [Google Scholar] [CrossRef] [PubMed]
  65. Wolter, T.; Wang, Y.; Hu, Q. Engineering Strategies to Mitigate Toxicities Associated with CAR-T Cell Therapy. BMEMat 2025, 3, e12109. [Google Scholar] [CrossRef]
  66. Lu, Y.; Zhao, F. Strategies to Overcome Tumor Relapse Caused by Antigen Escape After CAR T Therapy. Mol. Cancer 2025, 24, 126. [Google Scholar] [CrossRef]
  67. Zhai, Y.; Du, Y.; Li, G.; Yu, M.; Hu, H.; Pan, C.; Wang, D.; Shi, Z.; Yan, X.; Li, X.; et al. Trogocytosis of CAR Molecule Regulates CAR-T Cell Dysfunction and Tumor Antigen Escape. Nat. Commun. 2023, 14, 1708. [Google Scholar] [CrossRef]
  68. Dai, Z.; Mu, W.; Zhao, Y.; Cheng, J.; Lin, H.; Ouyang, K.; Jia, X.; Liu, J.; Wei, Q.; Wang, M.; et al. T Cells Expressing CD5/CD7 Bispecific Chimeric Antigen Receptors with Fully Human Heavy-Chain-Only Domains Mitigate Tumor Antigen Escape. Nat. Immunol. 2022, 23, 672–685. [Google Scholar] [CrossRef]
  69. Wei, J.; Mao, Z.; Wang, N.; Huang, L.; Cao, Y.; Sun, W.; Long, X.; Tan, J.; Li, C.; Xiao, Y.; et al. Long-Term Outcomes of Relapsed/Refractory Double-Hit Lymphoma (R/R DHL) Treated with CD19/22 CAR T-Cell Cocktail Therapy. Clin. Transl. Med. 2020, 10, e176. [Google Scholar] [CrossRef]
  70. Liu, Y.; Deng, B.; Hu, B.; Zhang, W.; Zhu, Q.; Liu, Y.; Wang, S.; Zhang, P.; Yang, Y.; Yang, J.; et al. Sequential Different B-Cell Antigen–Targeted CAR T-Cell Therapy for Pediatric Refractory/Relapsed Burkitt Lymphoma. Blood Adv. 2022, 6, 717–724. [Google Scholar] [CrossRef]
  71. Al-Haideri, M.; Tondok, S.B.; Safa, S.H.; Maleki, A.H. CAR-T Cell Combination Therapy: The Next Revolution in Cancer Treatment. J. Cancer Sci. Ther. 2022, 14, 445–452. [Google Scholar] [CrossRef]
  72. Liu, W.; Liu, W.; Zou, H.; Chen, L.; Huang, W.; Lv, R.; Xu, Y.; Liu, H.; Shi, Y.; Wang, K.; et al. Combinational therapy of CAR T-cell and HDT/ASCT demonstrates impressive clinical efficacy and improved CAR T-cell behavior in relapsed/refractory large B-cell lymphoma. J. Immunother. Cancer 2024, 12, e008857. [Google Scholar] [CrossRef] [PubMed]
  73. Wei, J.; Han, X.; Bo, J.; Han, W. Target Selection for CAR-T Therapy. J. Hematol. Oncol. 2019, 12, 43. [Google Scholar] [CrossRef]
  74. Schürch, C.M.; Rasche, L.; Frauenfeld, L.; Weinhold, N. A Review on Tumor Heterogeneity and Evolution in Multiple Myeloma: Pathological, Radiological, Molecular Genetics, and Clinical Integration. Virchows Arch. 2020, 476, 19–28. [Google Scholar] [CrossRef]
  75. Zhang, X.; Zhang, H.; Lan, H.; Wu, J.; Xiao, Y. CAR-T Cell Therapy in Multiple Myeloma: Current Limitations and Potential Strategies. Front. Immunol. 2023, 14, 1101495. [Google Scholar] [CrossRef]
  76. Shademan, B.; Karamad, V. CAR T Cells: Cancer Cell Surface Receptors are the Target for Cancer Therapy. Adv. Pharm. Bull. 2021, 12, 476–489. [Google Scholar] [CrossRef]
  77. Leow, C.C.Y.; Low, M.S.Y. Targeted Therapies for Multiple Myeloma. J. Pers. Med. 2021, 11, 334. [Google Scholar] [CrossRef] [PubMed]
  78. Labanieh, L.; Mackall, C.L. CAR Immune Cells: Design Principles, Resistance, and the Next Generation. Nature 2023, 614, 635–648. [Google Scholar] [CrossRef] [PubMed]
  79. Sterner, R.C.; Sterner, R.M. CAR-T Cell Therapy: Current Limitations and Potential Strategies. Blood Cancer J. 2021, 11, 81. [Google Scholar] [CrossRef]
  80. Available online: https://clinicaltrials.gov/study/NCT03361748 (accessed on 15 May 2025).
  81. Available online: https://clinicaltrials.gov/study/NCT04162210 (accessed on 15 May 2025).
  82. Available online: https://clinicaltrials.gov/study/NCT04555551 (accessed on 15 May 2025).
  83. He, X.; Zhang, X.; Yu, Z.; Meng, J.; Jiang, Y.; Zhao, Y.; Lyu, H.; Bai, X.; Xiao, X.; Zhao, M. Anti-FcRL5 CAR-T exhibits anti-MM activity against EMM after progression of anti-BCMA and anti-GPRC5D CAR-T: A case report. Blood Adv. 2025, 9, 1159–1162. [Google Scholar] [CrossRef]
  84. Available online: https://clinicaltrials.gov/study/NCT04351022 (accessed on 15 May 2025).
  85. Available online: https://clinicaltrials.gov/study/NCT03958656 (accessed on 15 May 2025).
  86. Available online: https://clinicaltrials.gov/study/NCT04430530 (accessed on 15 May 2025).
  87. Available online: https://clinicaltrials.gov/study/NCT02830724 (accessed on 15 May 2025).
  88. Available online: https://clinicaltrials.gov/study/NCT04623944 (accessed on 15 May 2025).
  89. Available online: https://clinicaltrials.gov/study/NCT02135406 (accessed on 15 May 2025).
  90. Available online: https://clinicaltrials.gov/study/NCT04318678 (accessed on 15 May 2025).
  91. Available online: https://clinicaltrials.gov/study/NCT02159495 (accessed on 15 May 2025).
  92. Available online: https://clinicaltrials.gov/study/NCT02623582 (accessed on 15 May 2025).
  93. Available online: https://clinicaltrials.gov/study/NCT04599543 (accessed on 15 May 2025).
  94. Available online: https://clinicaltrials.gov/study/NCT01716364 (accessed on 15 May 2025).
  95. Li, C.; Cao, W.; Que, Y.; Wang, Q.; Xiao, Y.; Gu, C.; Wang, D.; Wang, J.; Jiang, L.; Xu, H.; et al. A Phase I Study of Anti-BCMA CAR T Cell Therapy in Relapsed/Refractory Multiple Myeloma and Plasma Cell Leukemia. Cancer Med. 2021, 10, 346. [Google Scholar] [CrossRef]
  96. Tannoury, M.; Garnier, D.; Susin, S.A.; Bauvois, B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What’s Coming Next? Cancers 2022, 14, 6026. [Google Scholar] [CrossRef]
  97. Makarova, A.O.; Svirshchevskaya, E.V.; Titov, M.M. Prospects for the Use of Antibody-Drug Conjugates in Cancer Therapy. Russ. Chem. Rev. 2025, 94, 5597. [Google Scholar] [CrossRef]
  98. Sierro-Martínez, B.; Escamilla-Gómez, V. Next-generation BCMA-targeted Chimeric Antigen Receptor CARTemis-1: The Impact of Manufacturing Procedure on CAR T-cell Features. Cell. Oncol. 2025, 48, 219–237. [Google Scholar] [CrossRef] [PubMed]
  99. Abd-Aziz, N.; Poh, C.L. Development of Peptide-Based Vaccines for Cancer. J. Oncol. 2022, 2022, 9749363. [Google Scholar] [CrossRef] [PubMed]
  100. Roex, G.; Gordon, K.S.; Lion, E. Expanding the CAR Toolbox with High Throughput Screening Strategies for CAR Domain Exploration: A Comprehensive Review. J. Immunother. Cancer 2025, 13, e010658. [Google Scholar] [CrossRef] [PubMed]
  101. Utley, A.; Lipchick, B.; Lee, K.P.; Nikiforov, M.A. Targeting Multiple Myeloma Through the Biology of Long-lived Plasma Cells. Cancers 2020, 12, 2117. [Google Scholar] [CrossRef]
  102. Wang, Z.; Wang, M.; Wang, M.; Zhou, R. From Molecular Design to Clinical Translation: Dual-targeted CAR-T Strategies in Cancer Immunotherapy. Int. J. Biol. Sci. 2025, 21, 2676–2691. [Google Scholar] [CrossRef]
  103. Petrucci, M.T.; Bringhen, S.; Entrala Cerezo, C. Optimizing Treatment Sequencing in Multiple Myeloma: A Novel Model to Predict Survival Outcomes. Acta Haematol. 2024, 143, 2432815. [Google Scholar] [CrossRef]
  104. Krejci, M.; Adam, Z.; Krejci, M.; Pour, L.; Sandecka, V.; Stork, M. CAR-T cells for the treatment of relapsed/refractory multiple myeloma in 2022: Efficacy and toxicity. Neoplasma 2022, 69, 1008–1018. [Google Scholar] [CrossRef]
  105. Natrajan, K.; Kaushal, M.; George, B.; Kanapuru, B.; Theoret, M.R. FDA Approval Summary: Ciltacabtagene Autoleucel for Relapsed or Refractory Multiple Myeloma. Clin. Cancer Res. 2024, 30, 2865–2871. [Google Scholar] [CrossRef] [PubMed]
  106. Strassl, I.; Podar, K. The Preclinical Discovery and Clinical Development of Ciltacabtagene Autoleucel (Cilta-cel) for the Treatment of Multiple Myeloma. Expert Opin. Drug Discov. 2024, 19, 15–25. [Google Scholar] [CrossRef]
  107. Abebe, E.C.; Shiferaw, M.Y. Ciltacabtagene Autoleucel: The Second Anti-BCMA CAR T-Cell Therapeutic Armamentarium of Relapsed or Refractory Multiple Myeloma. Front. Immunol. 2022, 13, 991092. [Google Scholar] [CrossRef]
  108. van den Berg, J.; Holbro, A. EBMT-EHA 6th European CAR T-Cell Meeting 2024: Advancing Treatments and New Frontiers. Onco-Hema Heal. Times 2024, 20, 34–39. [Google Scholar] [CrossRef]
  109. Raab, M.S.; Cohen, Y.C.; Schjesvold, F.; Aardalen, K.; Oka, A.; Spencer, A.; Wermke, M.; Souza, A.D.; Kaufman, J.L.; Cafro, A.M.; et al. Preclinical Discovery and Initial Clinical Data of WVT078, a BCMA× CD3 Bispecific Antibody. Leukemia 2023, 37, 1735–1745. [Google Scholar] [CrossRef] [PubMed]
  110. Coffey, D.G.; Ataca Atilla, P.; Atilla, E.; Landgren, O. Single-Cell Analysis of the Multiple Myeloma Microenvironment after γ-Secretase Inhibition and CAR T-Cell Therapy. Blood 2025, 135, 1156–1168. [Google Scholar] [CrossRef]
  111. Yu, T.; Jiao, J.H.; Wu, M.F. CAR-T Cells in the Treatment of Multiple Myeloma: An Encouraging Cell Therapy. Front. Immunol. 2025, 16, 1499590. [Google Scholar] [CrossRef]
  112. Tang, C.; Zhang, Y. Potential alternatives to αβ-T cells to prevent graft-versus-host disease (GvHD) in allogeneic chimeric antigen receptor (CAR)-based cancer immunotherapy: A comprehensive review. Pathol Res Pract. 2024, 262, 155518. [Google Scholar] [CrossRef]
  113. Lonez, C.; Breman, E. Allogeneic CAR-T Therapy Technologies: Has the Promise Been Met? Cells 2024, 13, 146. [Google Scholar] [CrossRef]
  114. Ryszkiewicz, P.; Malinowska, B.; Schlicker, E. Polypharmacology: New Drugs in 2023–2024. Pharmacol. Rep. 2025, 77, 134–142. [Google Scholar] [CrossRef]
  115. Rodriguez-Otero, P.; van de Donk, N.W.C.J. GPRC5D as a Novel Target for the Treatment of Multiple Myeloma: A Narrative Review. Leukemia 2024, 38, 767–779. [Google Scholar] [CrossRef]
  116. Zhou, D.; Wang, Y.; Chen, C.; Li, Z.; Xu, K.; Zhao, K. Targeting GPRC5D for Multiple Myeloma Therapy. Leukemia 2024, 38, 1156–1168. [Google Scholar] [CrossRef] [PubMed]
  117. Yan, S.; Ming, X.; Zheng, R.; Zhu, X.; Xiao, Y. Application of GPRC5D Targeting Therapy in Relapsed Refractory Multiple Myeloma. Cancer Med. 2025, 14, 70764. [Google Scholar] [CrossRef]
  118. Nakashima, T.; Kagoya, Y. Current Progress of CAR-T-Cell Therapy for Patients with Multiple Myeloma. Int. J. Hematol. 2024, 109, 203–213. [Google Scholar] [CrossRef] [PubMed]
  119. Robat-Jazi, B.; Mahalleh, M.; Dashti, M.; Nejati, N.; Ahmadpour, M.; Alinejad, E.; Mohammadi, S.; Lorestani, P.; Hamidieh, A.A.; Habibi, M.A.; et al. A Systematic Review and Meta-analysis on the Safety and Efficacy of CAR T Cell Therapy Targeting GPRC5D in Patients with Multiple Myeloma: A New Insight in Cancer Immunotherapy. Anticancer Agents Med. Chem. 2025, 25, 1017–1028. [Google Scholar] [CrossRef]
  120. Zhang, M.; Wei, G.; Zhou, L.; Zhou, J.; Chen, S.; Zhang, W.; Wang, D.; Luo, X.; Cui, J.; Huang, S.; et al. GPRC5D CAR T cells (OriCAR-017) in patients with relapsed or refractory multiple myeloma (POLARIS): A first-in-human, single-center, single-arm, phase 1 trial. Lancet Haematol. 2023, 10, e107–e116. [Google Scholar] [CrossRef]
  121. Xia, J.; Sun, Q.; Zhou, D.; Li, H.; Wang, Y.; Qi, Y.; Cao, J.; Yan, Z.; Li, D.; Cheng, H.; et al. Anti-GPRC5D CAR T-cell therapy as a salvage treatment in patients with progressive multiple myeloma after anti-BCMA CAR T-cell therapy: A single-centre, single-arm, phase 2 trial. Lancet Haematol. 2025, 12, e365–e375. [Google Scholar] [CrossRef]
  122. Vera-Cruz, S.; Jornet Culubret, M.; Konetzki, V.; Alb, M. Cellular Therapies for Multiple Myeloma: Engineering Hope. Cancers 2024, 16, 3867. [Google Scholar] [CrossRef] [PubMed]
  123. Chang, C.W.; Luo, Y.; Pal, A.S.; Ding, H.; Lai, Y.; Guan, S.; Lan, W.; Li, W. A GPRC5D and BCMA bispecific CAR-T product demonstrated dual antigen targeting capability and efficacy against multiple myeloma cells bearing BCMA. J Immunother Cancer. 2024, 12 (Suppl. S2), A284. [Google Scholar] [CrossRef]
  124. Zhou, J.; Luo, X.; Wu, K.; Shi, F.; Lei, B.; Xu, Y.; Zhou, J.; He, X. Abstract 3183: GPRC5D and BCMA Bi-Specific CAR-T: Optimized CAR Design to Mitigate Antigen Escape and Elicit Deep and Durable Response in Heterogeneous Multiple Myeloma. Cancer Res. 2025, 85 (Suppl. S1), 3183. [Google Scholar] [CrossRef]
  125. Carlson, S.; Lin, T.L.; Larson, S.M. Allogeneic Chimeric Antigen Receptors (CARs) as an “Off-the-Shelf” Therapy in Multiple Myeloma. Immunotherapy 2025, 17, 249–260. [Google Scholar] [CrossRef] [PubMed]
  126. Wang, C.; Liu, T.; Wang, Q.; Gong, Y.; Gao, F.; Zhou, F.; Cao, Z. Abstract 6122: Off-the-Shelf BCMA/GPRC5D Dual Targeted CAR-NK Cell Therapy Combined with Daratumumab in Treating Multiple Myeloma. Cancer Res. 2025, 85 (Suppl. S1), 6122. [Google Scholar] [CrossRef]
  127. Yu, Z.; Li, H.; Lu, Q.; Zhang, Z.; Tong, A.; Niu, T. Fc Receptor-Like 5 (FCRL5)-Directed CAR-T Cells Exhibit Antitumor Activity Against Multiple Myeloma. Signal Transduct. Target. Ther. 2024, 9, 16. [Google Scholar] [CrossRef]
  128. Jiang, D.; Huang, H.; Qin, H.; Tang, K.; Shi, X.; Zhu, T.; Gao, Y.; Zhang, Y.; Tian, X.; Fu, J.; et al. Chimeric Antigen Receptor T Cells Targeting FcRH5 Provide Robust Tumour-Specific Responses in Murine Xenograft Models of Multiple Myeloma. Nat. Commun. 2023, 14, 3642. [Google Scholar] [CrossRef]
  129. Miller, K.C.; Hashmi, H.; Rajeeve, S. Beyond BCMA: The Next Wave of CAR T Cell Therapy in Multiple Myeloma. Front. Oncol. 2024, 14, 1398902. [Google Scholar] [CrossRef] [PubMed]
  130. Zhao, J.; Ren, Q.; Liu, X.; Guo, X.; Song, Y. Bispecific Antibodies Targeting BCMA, GPRC5D, and FcRH5 for Multiple Myeloma Therapy: Latest Updates from ASCO 2023 Annual Meeting. J. Hematol. Oncol. 2023, 16, 92. [Google Scholar] [CrossRef]
  131. Narra, R.K.; Peshin, S.; Dhakal, B. Novel Approaches of Cellular Therapy in Multiple Myeloma: Focus on Chimeric Antigen Receptor T-Cells. Acta Haematol. 2024, 148, 330–345. [Google Scholar] [CrossRef]
  132. Cohen, A.D.; Harrison, S.J.; Krishnan, A.; Fonseca, R. Initial Clinical Activity and Safety of BFCR4350A, a FcRH5/CD3 T-cell-Engaging Bispecific Antibody, in Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 746–754. [Google Scholar] [CrossRef]
  133. Waldschmidt, J.M.; Rasche, L.; Kortüm, K.M. Bispecific Antibody Constructs in Multiple Myeloma: Affinities, Dosing Strategies, and Future Perspectives. Clin. Lymphoma Myeloma Leuk. 2024, 24, 267–277. [Google Scholar] [CrossRef]
  134. O’Neill, N.; van de Donk, N.W.C.J. T-Cell Redirecting Bispecific Antibodies in Multiple Myeloma: Current Landscape and Future Directions. EJHaem 2023, 4, 811–822. [Google Scholar] [CrossRef]
  135. He, Y.; Vlaming, M.; van Meerten, T.; Bremer, E. The Implementation of TNFRSF Co-stimulatory Domains in CAR-T Cells for Optimal Functional Activity. Cancers 2022, 14, 299. [Google Scholar] [CrossRef] [PubMed]
  136. Honikel, M.M.; Olejniczak, S.H. Co-Stimulatory Receptor Signaling in CAR-T Cells. Biomolecules 2022, 12, 1303. [Google Scholar] [CrossRef]
  137. Fu, B.; Liu, R.; Gao, G.; Lin, Z.; He, A. Mechanisms and Salvage Treatments in Patients with Multiple Myeloma Relapsed Post-BCMA CAR-T Cell Therapy. Front. Immunol. 2024, 15, 1433774. [Google Scholar] [CrossRef]
  138. Mishra, A.K.; Gupta, A.; Dagar, G.; Das, D.; Chakraborty, A.; Haque, S.; Prasad, C.P.; Singh, A.; Bhat, A.A.; Macha, M.A.; et al. CAR-T-Cell Therapy in Multiple Myeloma: B-Cell Maturation Antigen (BCMA) and Beyond. Vaccines 2023, 11, 1721. [Google Scholar] [CrossRef] [PubMed]
  139. Lee, H.C.; Deng, Q.W.; Zhao, Y.J. The Calcium Signaling Enzyme CD38—A Paradigm for Membrane Topology Defining Distinct Protein Functions. Cell Calcium 2022, 99, 102424. [Google Scholar] [CrossRef]
  140. Adolph, T.E.; Meyer, M.; Jukic, A.; Tilg, H. Heavy Arch: From Inflammatory Bowel Diseases to Metabolic Disorders. Gut 2024, 73, 1376–1385. [Google Scholar] [CrossRef] [PubMed]
  141. Kim, B.J.; Choi, Y.M.; Rah, S.Y.; Park, D.R.; Park, S.A.; Chung, Y.J.; Park, S.M.; Park, J.K.; Jang, K.Y.; Kim, U.H. Seminal CD38 Is a Pivotal Regulator for Fetomaternal Tolerance. Proc. Natl. Acad. Sci. USA 2015, 112, 1559–1564. [Google Scholar] [CrossRef]
  142. Encinas, C.; Lozano, V.; Hlavacek, P.; Llinares, J.; Toribio-Castelló, S.; Carcedo, D.; Asís-Montalt, J.; Martínez-López, J. Cost-Effectiveness of Elranatamab Versus Current Therapies for the Management of Patients with Triple-Class Exposed, Relapsed and Refractory Multiple Myeloma, Including Other Bispecific and Physician’s Choice of Treatment in Spain. Oncol. Ther. 2025, 13, 381–395. [Google Scholar] [CrossRef]
  143. Hashmi, H.; Husnain, M.; Khan, A. CD38-Directed Therapies for Management of Multiple Myeloma. Immunotherapy 2021, 13, 273–285. [Google Scholar] [CrossRef]
  144. García-Guerrero, E.; Sierro-Martínez, B. Overcoming Chimeric Antigen Receptor (CAR) Modified T-Cell Therapy Limitations in Multiple Myeloma. Front. Immunol. 2020, 11, 1128. [Google Scholar] [CrossRef]
  145. Deng, H.; Wang, Q.; Tong, X.; Cui, Z. Recent Advances of CAR-T Cells in Acute Myeloid Leukemia. Ther. Adv. Hematol. 2025, 16, 1–19. [Google Scholar] [CrossRef] [PubMed]
  146. Peng, Z.; Wang, J.; Guo, J.; Li, X.; Wang, S.; Xie, Y.; Jiang, H.; Wang, Y.; Wang, M.; Hu, M.; et al. All-Trans Retinoic Acid Improves NSD2-Mediated RARα Phase Separation and Efficacy of Anti-CD38 CAR T-Cell Therapy in Multiple Myeloma. J. Immunother. Cancer 2023, 11, e006325. [Google Scholar] [CrossRef] [PubMed]
  147. Sheykhhasan, M.; Ahmadieh-Yazdi, A. CAR T Therapies in Multiple Myeloma: Unleashing the Future. Cancer Gene Ther. 2024, 5, 210–222. [Google Scholar] [CrossRef]
  148. Keller, A.L.; Reiman, L.T.; Perez de Acha, O.; Parzych, S.E.; Forsberg, P.A.; Kim, P.S.; Bisht, K.; Wang, H.; Van de Velde, H.; Sherbenou, D.W.; et al. Ex Vivo Efficacy of SAR442257 Anti-CD38 Trispecific T-cell Engager in Multiple Myeloma Relapsed After Daratumumab and BCMA-targeted Therapies. Cancer Res Commun. 2024, 4, 757–764. [Google Scholar] [CrossRef] [PubMed]
  149. Mei, H.; Li, C.; Jiang, H.; Zhao, X.; Huang, Z.; Jin, D.; Guo, T.; Kou, H.; Liu, L.; Tang, L.; et al. A Bispecific CAR-T Cell Therapy Targeting BCMA and CD38 in Relapsed or Refractory Multiple Myeloma. Leukemia 2021, 35, 667–678. [Google Scholar] [CrossRef]
  150. De Novellis, D.; Fontana, R.; Giudice, V.; Serio, B. Innovative Anti-CD38 and Anti-BCMA Targeted Therapies in Multiple Myeloma: Mechanisms of Action and Resistance. Int. J. Mol. Sci. 2022, 24, 645. [Google Scholar] [CrossRef]
  151. Bruno, B.; Wäsch, R.; Engelhardt, M.; Gay, F. European Myeloma Network Perspective on CAR T-Cell Therapies for Multiple Myeloma. J. Clin. Oncol. 2021, 39, 2681–2692. [Google Scholar] [CrossRef]
  152. Zamagni, E.; Tacchetti, P.; Pantani, L.; Cavo, M. Anti-CD38 and Anti-SLAMF7: The Future of Myeloma Immunotherapy. Crit. Rev. Oncol. Hematol. 2018, 132, 131–142. [Google Scholar] [CrossRef]
  153. Veillette, A.; Guo, H. CS1, a SLAM Family Receptor Involved in Immune Regulation, Is a Therapeutic Target in Multiple Myeloma. Crit. Rev. Oncol. Hematol. 2013, 88, 168–177. [Google Scholar] [CrossRef]
  154. Zhang, Z.; Zhang, Y.; Chen, Z.; Xia, L. Emerging Roles of SLAMF7 in Immune Cells and Related Diseases. Innate Immun. 2025, 31, 267–279. [Google Scholar] [CrossRef]
  155. Gogishvili, T.; Danhof, S.; Prommersberger, S.; Rydzek, J.; Schreder, M.; Brede, C.; Einsele, H.; Hudecek, M. SLAMF7-CAR T Cells Eliminate Myeloma and Confer Selective Fratricide of SLAMF7+ Normal Lymphocytes. Blood 2017, 130, 2838–2847. [Google Scholar] [CrossRef] [PubMed]
  156. Kostelecka, K.; Brylinski, L.; Wolinski, F.; Smyk, M. The Potential Role of CAR-T in Multiple Myeloma Treatment: A Review. Cent. Eur. J. Oncol. 2024, 12, 133–144. [Google Scholar] [CrossRef]
  157. Wang, H.; Pan, W. Challenges of Chimeric Antigen Receptor-T/Natural Killer Cell Therapy in the Treatment of Solid Tumors: Focus on Colorectal Cancer and Evaluation of Combination Therapies. Mol. Cell. Biochem. 2023, 478, 967–980. [Google Scholar] [CrossRef] [PubMed]
  158. Prommersberger, S.; Reiser, M.; Beckmann, J.; Danhof, S.; Amberger, M.; Quade-Lyssy, P.; Einsele, H.; Hudecek, M.; Bonig, H.; Ivics, Z. CARAMBA: A First-in-Human Clinical Trial with SLAMF7 CAR-T Cells Prepared by Virus-Free Sleeping Beauty Gene Transfer to Treat Multiple Myeloma. Gene Ther. 2021, 28, 738–746. [Google Scholar] [CrossRef] [PubMed]
  159. Chu, E.; Wu, J.; Kang, S.S.; Kang, Y. SLAMF7 as a Promising Immunotherapeutic Target in Multiple Myeloma Treatments. Curr. Oncol. 2023, 30, 573. [Google Scholar] [CrossRef]
  160. Hassan, N.; Efing, J.; Kiesel, L.; Bendas, G.; Götte, M. The Tissue Factor Pathway in Cancer: Overview and Role of Heparan Sulfate Proteoglycans. Cancers 2023, 15, 1524. [Google Scholar] [CrossRef]
  161. Steiner, N.; Gunsilius, E. CAR-T Cells in Multiple Myeloma: Current Status. Memo—Mag. Eur. Med. Oncol. 2020, 13, 158–165. [Google Scholar] [CrossRef]
  162. van de Donk, N.W.C.J.; Usmani, S.Z.; Yong, K. CAR T-Cell Therapy for Multiple Myeloma: State of the Art and Prospects. Lancet Haematol. 2021, 8, e446–e461. [Google Scholar] [CrossRef]
  163. Ding, L.; Hu, Y.; Huang, H. Novel Progresses of Chimeric Antigen Receptor (CAR) T Cell Therapy in Multiple Myeloma. Stem Cell Investig. 2021, 8, 33–45. [Google Scholar] [CrossRef]
  164. Riccardi, F.; Tangredi, C.; Dal Bo, M.; Toffoli, G. Targeted Therapy for Multiple Myeloma: An Overview on CD138-Based Strategies. Front. Oncol. 2024, 14, 1370854. [Google Scholar] [CrossRef]
  165. Kasap, C.; Izgutdina, A.; Patiño-Escobar, B.; Kang, A. Targeting High-Risk Multiple Myeloma Genotypes with Optimized Anti-CD70 CAR-T Cells. Nat. Commun. 2024, 15, 1234. [Google Scholar] [CrossRef]
  166. Kasap, C.; Izgutdina, A.; Patiño Escobar, B.; Lin, Y.-T.; Geng, H.; Wicaksono, G.; Dupere-Richer, D.; Ramos, E.; Barpanda, A.; Vijayanarayanan, A.; et al. Discovery and Development of CD70 as a Cellular Therapy Target in High-Risk Multiple Myeloma. Blood 2023, 142 (Suppl. S1), 465. [Google Scholar] [CrossRef]
  167. Sauer, T.; Parikh, K.; Sharma, S.; Omer, B. CD70-Specific CAR T Cells Have Potent Activity Against Acute Myeloid Leukemia Without HSC Toxicity. Blood 2021, 138, 318–330. [Google Scholar] [CrossRef]
  168. Wu, G.; Guo, S.; Luo, Q.; Wang, X.; Deng, W.; Ouyang, G.; Pu, J.J.; Lei, W.; Qian, W. Preclinical Evaluation of CD70-Specific CAR T Cells Targeting Acute Myeloid Leukemia. Front. Immunol. 2023, 14, 1093750. [Google Scholar] [CrossRef] [PubMed]
  169. Pal, S.K.; Reimers, M.A.; Garmezy, B.; Xu, W.; Hoimes, C.J.; Weinstein, M.; Cullingford, E.L.; Ma, A.; Williams, L.M.; Dar, H.; et al. A Phase 1/2, Open-Label, Multicenter, Dose Escalation and Cohort Expansion Study of the Safety and Efficacy of Anti-CD70 Allogeneic CRISPR-Cas9–Engineered T Cells (CTX131) in Adult Patients with Relapsed or Refractory Solid Tumors. J. Clin. Oncol. 2024, 42 (Suppl. S15), TPS2676. [Google Scholar] [CrossRef]
  170. Iyer, S.P.; Sica, R.A.; Ho, P.J.; Prica, A.; Zain, J.; Foss, F.M.; Hu, B.; Beitinjaneh, A.; Weng, W.K.; Kim, Y.H.; et al. Safety and Activity of CTX130, a CD70-Targeted Allogeneic CRISPR-Cas9-Engineered CAR T-Cell Therapy, in Patients with Relapsed or Refractory T-Cell Malignancies (COBALT-LYM): A Single-Arm, Open-Label, Phase 1, Dose-Escalation Study. Lancet Oncol. 2024, 25, 110–122. [Google Scholar] [CrossRef]
  171. Lin, P.; Silva, F.C.R.; Gilbert, A.L.; Acharya, S. CD70 CAR NK Cells in the Treatment of Multiple Myeloma. Blood 2023, 142, 3463. [Google Scholar] [CrossRef]
  172. Guo, S.; Lei, W.; Jin, X.; Liu, H.; Wang, J.Q.; Deng, W.; Qian, W. CD70-Specific CAR NK Cells Expressing IL-15 for the Treatment of CD19-Negative B-Cell Malignancy. Blood Adv. 2024, 8, 2635–2645. [Google Scholar] [CrossRef]
  173. Testa, U.; Castelli, G.; Pelosi, E. Emerging Role of Chimeric Antigen Receptor-Natural Killer Cells for the Treatment of Hematologic Malignancies. Cancers 2025, 17, 1454. [Google Scholar] [CrossRef]
  174. Olejarz, W.; Basak, G. Emerging Therapeutic Targets and Drug Resistance Mechanisms in Immunotherapy of Hematological Malignancies. Cancers 2023, 15, 5765. [Google Scholar] [CrossRef]
  175. Boneva, E.; Shivarov, V.; Ivanova, M. A Concise Review of the Role of the NKG2D Receptor and Its Ligands in Cancer. Immuno 2025, 5, 9. [Google Scholar] [CrossRef]
  176. Tan, G.; Spillane, K.M.; Maher, J. The Role and Regulation of the NKG2D/NKG2D Ligand System in Cancer. Biology 2023, 12, 1079. [Google Scholar] [CrossRef]
  177. Prajapati, K.; Perez, C.; Rojas, L.B.P.; Burke, B.; Guevara-Patino, J.A. Functions of NKG2D in CD8+ T Cells: An Opportunity for Immunotherapy. Cell. Mol. Immunol. 2018, 15, 470–479. [Google Scholar] [CrossRef]
  178. Dosani, T.; Carlsten, M.; Maric, I.; Landgren, O. The Cellular Immune System in Myelomagenesis: NK Cells and T Cells in the Development of Myeloma and Their Uses in Immunotherapies. Blood Cancer J. 2015, 5, e306. [Google Scholar] [CrossRef] [PubMed]
  179. Liu, J.; Dai, K.; Saliu, M.A.; Salisu, M.D.; Gan, J.; Afolabi, L.O.; Yan, D.; Zhang, G.; Liu, M.; Wan, X.; et al. Sodium valproate enhances efficacy of NKG2D CAR-T cells against glioblastoma. Front. Immunol. 2025, 15, 1519777. [Google Scholar] [CrossRef]
  180. Trabolsi, A.; Arumov, A.; Schatz, J.H. Bispecific Antibodies and CAR-T Cells: Dueling Immunotherapies for Large B-Cell Lymphomas. Blood Cancer J. 2024, 14, 27. [Google Scholar] [CrossRef] [PubMed]
  181. Dada, R. Redefining Precision Management of R/R Large B-Cell Lymphoma: Novel Antibodies Take on CART and BMT in the Quest for Future Treatment Strategies. Cells 2023, 12, 1858. [Google Scholar] [CrossRef]
  182. Moreno-Cortes, E.; Forero-Forero, J.V. Chimeric Antigen Receptor T Cell Therapy in Oncology–Pipeline at a Glance: Analysis of the ClinicalTrials.gov Database. Leuk. Res. 2021, 104, 106567. [Google Scholar] [CrossRef]
  183. Teoh, P.J.; Chng, W.J. CAR T-Cell Therapy in Multiple Myeloma: More Room for Improvement. Blood Cancer J. 2021, 11, 84. [Google Scholar] [CrossRef]
  184. Rejeski, K.; Jain, M.D.; Smith, E.L. Mechanisms of Resistance and Treatment of Relapse After CAR T-Cell Therapy for Large B-Cell Lymphoma and Multiple Myeloma. Transplant. Cell Ther. 2023, 29, 418–428. [Google Scholar] [CrossRef]
  185. Rubinstein, P.G.; Galvez, C.; Ambinder, R.F. Hematopoietic Stem Cell Transplantation and Cellular Therapy in Persons Living with HIV. Curr. Opin. Infect. Dis. 2024, 37, 254–263. [Google Scholar] [CrossRef] [PubMed]
  186. Almotiri, A. CAR T-Cell Therapy in Acute Myeloid Leukemia. Saudi Med. J. 2024, 45, 1007–1019. [Google Scholar] [CrossRef]
  187. Salehi-Shadkami, H.; Sedghi, M.; Tavoosi, S. Effective Targeting of CD19 Positive Primary B-ALL Cells Using CAR-NK Cells Generated with mRNA-LNPs. bioRxiv 2024. [Google Scholar] [CrossRef]
  188. Kang, L.; Zhang, J.; Li, M.; Xu, N.; Qi, W.; Tan, J.; Lou, X.; Yu, Z.; Sun, J.; Wang, Z.; et al. Characterization of Novel Dual Tandem CD19/BCMA Chimeric Antigen Receptor T Cells to Potentially Treat Multiple Myeloma. Biomark. Res. 2020, 8, 14. [Google Scholar] [CrossRef]
  189. Jiang, H.; Dong, B.; Gao, L.; Liu, L.; Ge, J.; He, A.; Du, J.; Li, L.; Lu, J.; Chen, X.; et al. Clinical Results of a Multicenter Study of the First-in-Human Dual BCMA and CD19 Targeted Novel Platform Fast CAR-T Cell Therapy for Patients with Relapsed/Refractory Multiple Myeloma. Blood 2020, 136 (Suppl. S1), 25–26. [Google Scholar] [CrossRef]
  190. Du, J.; Fu, W.; Jiang, H.; Dong, B.; Gao, L.; Liu, L.; Ge, J.; He, A.; Li, L.; Lu, J.; et al. P869: Updated Results of a Phase I, Open-Label Study of BCMA/CD19 Dual-Targeting FasTCAR-T GC012F for Patients with Relapsed/Refractory Multiple Myeloma (RRMM). HemaSphere 2023, 7 (Suppl. S3), e84060bf. [Google Scholar] [CrossRef]
  191. Steinhardt, M.; Einsele, H. Advances in CD19-Targeting CAR-T Cell Therapies for Multiple Myeloma. Expert Opin. Biol. Ther. 2025, 25, 229–240. [Google Scholar] [CrossRef]
  192. Xu, H.; Guan, C.; Xu, P.; Zhou, D.; Xu, Y.; Chen, B.; Bai, H. Clinical Efficacy and Safety of Combined Anti-BCMA and Anti-CD19 CAR-T Cell Therapy for Relapsed/Refractory Multiple Myeloma: A Systematic Review and Meta-Analysis. Front. Oncol. 2024, 14, 1355643. [Google Scholar] [CrossRef]
  193. Shi, X.; Yan, L.; Shang, J.; Kang, L.; Yan, Z.; Jin, S.; Zhu, M.; Chang, H.; Gong, F.; Zhou, J.; et al. Anti-BCMA CAR T Cell Therapy Followed by Lenalidomide Maintenance after Autologous Stem-Cell Transplantation for High-Risk Newly Diagnosed Multiple Myeloma. Am. J. Hematol. 2022, 97, 238–246. [Google Scholar] [CrossRef]
  194. D’Agostino, M.; Raje, N. Anti-BCMA CAR T-Cell Therapy in Multiple Myeloma: Can We Do Better? Leukemia 2020, 34, 21–34. [Google Scholar] [CrossRef]
  195. Shi, M.; Wang, J.; Huang, H.; Liu, D.; Cheng, H. Bispecific CAR T Cell Therapy Targeting BCMA and CD19 in Relapsed/Refractory Multiple Myeloma: A Phase I/II Trial. Nat. Commun. 2024, 15, 1234–1244. [Google Scholar] [CrossRef] [PubMed]
  196. Pinti, M.; Gibellini, L.; Lo Tartaro, D.; De Biasi, S.; Nasi, M.; Borella, R.; Fidanza, L.; Neroni, A.; Troiano, L.; Franceschi, C.; et al. A Comprehensive Analysis of Cytokine Network in Centenarians. Int. J. Mol. Sci. 2023, 24, 2719. [Google Scholar] [CrossRef] [PubMed]
  197. Hu, Y.; Xie, Y.; Wang, X.; Yang, L.; Geng, H.; Yi, Z.; Zhang, Y.; Ma, L.; Chen, F. Targeting BCMA in Multiple Myeloma: Designs, Challenges, and Future Directions. Cancer Immunol. Immunother. 2025, 74, 77. [Google Scholar] [CrossRef]
  198. Larson, R.C.; Castano, A.; Bouffard, A.A.; Kann, M.C.; Schmidts, A.; Gallagher, K.M.; Maus, M.V. Abstract 556: Novel anti-TACI single and dual-targeting CAR T cells overcome BCMA antigen loss in multiple myeloma. Cancer Res. 2022, 82 (Suppl. S12), 556. [Google Scholar] [CrossRef]
  199. Ohmine, K.; Uchibori, R. Novel Immunotherapies in Multiple Myeloma. Int. J. Hematol. 2022, 115, 799–810. [Google Scholar] [CrossRef]
  200. Larson, R.C.; Kann, M.C.; Graham, C.; Mount, C.W.; Castano, A.P.; Lee, W.-H.; Bouffard, A.A.; Takei, H.N.; Almazan, A.J.; Scarfó, I.; et al. Anti-TACI Single and Dual-Targeting CAR T Cells Overcome BCMA Antigen Loss in Multiple Myeloma. Nat. Commun. 2023, 14, 7509. [Google Scholar] [CrossRef]
  201. Cornelis, R.; Chang, H.D.; Radbruch, A. Keeping Up with the Stress of Antibody Production: BAFF and APRIL Maintain Memory Plasma Cells. Curr. Opin. Immunol. 2021, 71, 97–102. [Google Scholar] [CrossRef]
  202. Eslami, M.; Schuepbach-Mallepell, S.; Diana, D.; Willen, L.; Kowalczyk-Quintas, C.; Desponds, C.; Peter, B.; Vigolo, M.; Renevey, F.; Donzé, O.; et al. Unique and Redundant Roles of Mouse BCMA, TACI, BAFF, APRIL, and IL-6 in Supporting Antibody-Producing Cells in Different Tissues. Proc. Natl. Acad. Sci. USA 2024, 121, e2404309121. [Google Scholar] [CrossRef]
  203. Shah, N.; Chari, A.; Scott, E.; Mezzi, K.; Usmani, S.Z. B-Cell Maturation Antigen (BCMA) in Multiple Myeloma: Rationale for Targeting and Current Therapeutic Approaches. Leukemia 2020, 34, 985–1005. [Google Scholar] [CrossRef]
  204. Camviel, N.; Wolf, B.; Croce, G.; Gfeller, D. Both APRIL and Antibody-Fragment-Based CAR T Cells for Myeloma Induce BCMA Downmodulation by Trogocytosis and Internalization. Front. Immunol. 2022, 13, 1002500. [Google Scholar] [CrossRef]
  205. Ullah, M.; Mackay, F. The BAFF-APRIL System in Cancer. Cancers 2023, 15, 1791. [Google Scholar] [CrossRef] [PubMed]
  206. Schmidts, A.; Ormhøj, M.; Choi, B.D.; Taylor, A.O.; Bouffard, A.A.; Scarfò, I.; Larson, R.C.; Frigault, M.J.; Gallagher, K.; Castano, A.P.; et al. Rational Design of a Trimeric APRIL-Based CAR-Binding Domain Enables Efficient Targeting of Multiple Myeloma. Blood Adv. 2019, 3, 324–334. [Google Scholar] [CrossRef] [PubMed]
  207. Medler, J.; Wajant, H. Tumor Necrosis Factor Receptor-2 (TNFR2): An Overview of an Emerging Drug Target. Expert Opin. Ther. Targets 2019, 23, 295–307. [Google Scholar] [CrossRef] [PubMed]
  208. Li, F.; Gao, C.; Huang, Y.; Qiao, Y.; Xu, H.; Liu, S.; Wu, H. Unraveling the Breast Cancer Tumor Microenvironment: Crucial Factors Influencing Natural Killer Cell Function and Therapeutic Strategies. Int. J. Biol. Sci. 2025, 21, 2606–2628. [Google Scholar] [CrossRef]
  209. Yang, Y.; Islam, M.S.; Hu, Y.; Chen, X. TNFR2: Role in Cancer Immunology and Immunotherapy. ImmunoTargets Ther. 2021, 10, 103–122. [Google Scholar] [CrossRef]
  210. Li, L.; Ye, R.; Li, Y.; Pan, H.; Han, S.; Lu, Y. Targeting TNFR2 for Cancer Immunotherapy: Recent Advances and Future Directions. J. Transl. Med. 2024, 22, 812. [Google Scholar] [CrossRef]
  211. Moatti, A.; Cohen, J.L. The TNF-α/TNFR2 Pathway: Targeting a Brake to Release the Anti-Tumor Immune Response. Front. Cell Dev. Biol. 2021, 9, 725473. [Google Scholar] [CrossRef]
  212. Wu, Y.; Chen, D.; Gao, Y.; Xu, Q.; Zhou, Y.; Ni, Z.; Na, M. Immunosuppressive Regulatory Cells in Cancer Immunotherapy: Restrain or Modulate? Hum. Cell 2024, 37, 931–943. [Google Scholar] [CrossRef]
  213. Ba, H.; Dai, Z.; Zhang, Z.; Zhang, P.; Yin, B.; Wang, J.; Li, Z.; Zhou, X. Antitumor Effect of CAR-T Cells Targeting Transmembrane Tumor Necrosis Factor Alpha Combined with PD-1 mAb on Breast Cancers. Front. Immunol. 2023, 11, e003837. [Google Scholar] [CrossRef]
  214. Takahashi, H.; Yoshimatsu, G.; Faustman, D.L. The Roles of TNFR2 Signaling in Cancer Cells and the Tumor Microenvironment and the Potency of TNFR2 Targeted Therapy. Cells 2022, 11, 1952. [Google Scholar] [CrossRef]
  215. Dawson, N.A.J.; Rosado-Sánchez, I.; Novakovsky, G.E.; Fung, V.C.W.; Huang, Q.; McIver, E.; Sun, G.; Gillies, J.; Speck, M.; Orban, P.C.; et al. Functional Effects of Chimeric Antigen Receptor Co-Receptor Signaling Domains in Human Regulatory T Cells. Sci. Transl. Med. 2020, 12, eaaz3866. [Google Scholar] [CrossRef] [PubMed]
  216. D’Accardo, C.; Porcelli, G.; Mangiapane, L.R.; Modica, C.; Pantina, V.D.; Roozafzay, N.; Di Franco, S.; Gaggianesi, M.; Veschi, V.; Lo Iacono, M.; et al. Cancer cell targeting by CAR-T cells: A matter of stemness. Front. Mol. Med. 2022, 2, 1055028. [Google Scholar] [CrossRef] [PubMed]
  217. Casucci, M.; Nicolis di Robilant, B.; Falcone, L.; Camisa, B.; Norelli, M.; Genovese, P.; Gentner, B.; Gullotta, F.; Ponzoni, M.; Bernardi, M.; et al. CD44v6-Targeted T Cells Mediate Potent Antitumor Effects Against Acute Myeloid Leukemia and Multiple Myeloma. Blood 2013, 122, 3461–3472. [Google Scholar] [CrossRef]
  218. Wang, Z.; Zhao, K.; Hackert, T.; Zöller, M. CD44/CD44v6: A Reliable Companion in Cancer-Initiating Cell Maintenance and Tumor Progression. Front. Cell Dev. Biol. 2018, 6, 97. [Google Scholar] [CrossRef] [PubMed]
  219. George, L.L.; Deshpande, S.R.; Cortese, M.J.; Kendall, E.K.; Chattaraj, A.; Shah, Z.; Zhao, J.; Anwer, F. Emerging Targets and Cellular Therapy for Relapsed Refractory Multiple Myeloma: A Systematic Review. Clin. Lymphoma Myeloma Leuk. 2021, 21, 741–751. [Google Scholar] [CrossRef]
  220. Gupta, S.; Master, S.; Graham, C. Extramedullary Multiple Myeloma: A Patient-Focused Review of the Pathogenesis of Bone Marrow Escape. World J. Oncol. 2022, 13, 311–319. [Google Scholar] [CrossRef]
  221. Rodríguez-Lobato, L.G.; Ganzetti, M.; Fernández de Larrea, C.; Hudecek, M.; Einsele, H.; Danhof, S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front. Oncol. 2020, 10, 1243. [Google Scholar] [CrossRef]
  222. Lu, L.; Xie, M.; Yang, B.; Zhao, W.B.; Cao, J. Enhancing the Safety of CAR-T Cell Therapy: Synthetic Genetic Switch for Spatiotemporal Control. Sci. Adv. 2024, 10, eadj6251. [Google Scholar] [CrossRef]
  223. Ciulean, S.I.; Fischer, J.; Quaiser, A.; Bach, C.; Abken, H.; Tretbar, U.S.; Fricke, S.; Köhl, U.; Schmiedel, D.; Grunwald, T. Harnessing anti-CD44v6 CAR-NK cells for effective head and neck squamous cell carcinoma treatment. J. Immunother. Cancer 2024, 12 (Suppl. S2), A250. [Google Scholar] [CrossRef]
  224. Ciulean, I.S.; Fischer, J.; Quaiser, A.; Bach, C.; Abken, H.; Tretbar, U.S.; Fricke, S.; Koehl, U.; Schmiedel, D.; Grunwald, T.; et al. CD44v6 Specific CAR-NK Cells for Targeted Immunotherapy of Head and Neck Squamous Cell Carcinoma. Front. Immunol. 2023, 14, 1290488. [Google Scholar] [CrossRef]
  225. Porcellini, S.; Asperti, C.; Corna, S.; Cicoria, E.; Valtolina, V.; Stornaiuolo, A.; Valentinis, B.; Bordignon, C.; Traversari, C. CAR T Cells Redirected to CD44v6 Control Tumor Growth in Lung and Ovary Adenocarcinoma Bearing Mice. Front. Immunol. 2020, 11, 99. [Google Scholar] [CrossRef] [PubMed]
  226. Lodewijk, I.; Dueñas, M.; Paramio, J.M. CD44v6, STn & O-GD2: Promising Tumor Associated Antigens Paving the Way for New Targeted Cancer Therapies. Front. Immunol. 2023, 14, 1272681. [Google Scholar] [CrossRef]
  227. Carrabba, M.G.; Casucci, M.; Hudecek, M.; Quintarelli, C.; Briones, J.; Hajek, R.; Sierra, J.; Locatelli, F.; Einsele, H.; Bordignon, C.; et al. Phase I–IIa Clinical Trial to Assess Safety and Efficacy of MLM-CAR44.1, a CD44v6 Directed CAR-T in Relapsed/Refractory Acute Myeloid Leukemia (AML) and Multiple Myeloma (MM). Blood 2018, 132 (Suppl. S1), 5790. [Google Scholar] [CrossRef]
  228. Van der Schans, J.J.; Van de Donk, N.W.C.J.; Mutis, T. Dual Targeting to Overcome Current Challenges in Multiple Myeloma CAR T-Cell Treatment. Front. Oncol. 2020, 10, 1362. [Google Scholar] [CrossRef] [PubMed]
  229. Hosen, N.; Matsunaga, Y.; Hasegawa, K.; Matsuno, H.; Nakamura, Y.; Makita, M.; Watanabe, K.; Yoshida, M.; Satoh, K.; Morimoto, S.; et al. The Activated Conformation of Integrin β7 Is a Novel Multiple Myeloma–Specific Target for CAR T Cell Therapy. Nat. Med. 2017, 23, 1436–1443. [Google Scholar] [CrossRef]
  230. Ritchie, D.S.; Neeson, P.J.; Khot, A.; Peinert, S.; Tai, T.; Tainton, K.; Chen, K.; Shin, M.; Wall, D.M.; Hönemann, D.; et al. Persistence and Efficacy of Second Generation CAR T Cell Against the LeY Antigen in Acute Myeloid Leukemia. Mol. Ther. 2013, 21, 2122–2129. [Google Scholar] [CrossRef]
  231. Savani, M.; Oluwole, O.; Dholaria, B. New Targets for CAR T Therapy in Hematologic Malignancies. Semin. Hematol. 2021, 58, 208–215. [Google Scholar] [CrossRef]
  232. Cottini, F.; Rodriguez, J.; Hughes, T.; Sharma, N.; Guo, L.; Lozanski, G.; Liu, B.; Cocucci, E.; Yang, Y.; Benson, D.; et al. Redefining CD56 as a Biomarker and Therapeutic Target in Multiple Myeloma. Mol. Cancer Res. 2022, 20, 1083–1095. [Google Scholar] [CrossRef]
  233. Koumpis, E.; Tassi, I.; Malea, T.; Papathanasiou, K.; Papakonstantinou, I.; Serpanou, A.; Tsolas, E.; Kapsali, E.; Vassilakopoulos, T.P.; Papoudou-Bai, A.; et al. CD56 Expression in Multiple Myeloma: Correlation with Poor Prognostic Markers but No Effect on Outcome. Pathol. Res. Pract. 2021, 225, 153567. [Google Scholar] [CrossRef]
  234. Sahara, N.; Takeshita, A. Prognostic Significance of Surface Markers Expressed in Multiple Myeloma: CD56 and Other Antigens. Leuk. Lymphoma 2004, 45, 61–65. [Google Scholar] [CrossRef]
  235. Hosen, N. Chimeric Antigen Receptor T-Cell Therapy for Multiple Myeloma. Int. J. Hematol. 2020, 112, 62–67. [Google Scholar] [CrossRef] [PubMed]
  236. Uckun, F.M. Dual targeting of multiple myeloma stem cells and myeloid-derived suppressor cells for treatment of chemotherapy-resistant multiple myeloma. Front. Oncol. 2021, 11, 760382. [Google Scholar] [CrossRef]
  237. Ho, M.; Xiao, A.; Yi, D.; Zanwar, S.; Bianchi, G. Treating multiple myeloma in the context of the bone marrow microenvironment. Curr. Oncol. 2022, 29, 8975–9005. [Google Scholar] [CrossRef] [PubMed]
  238. Testa, U.; Pelosi, E.; Castelli, G. CD123 as a therapeutic target in the treatment of hematological malignancies. Cancers 2019, 11, 1358. [Google Scholar] [CrossRef] [PubMed]
  239. Caruso, S.; De Angelis, B.; Del Bufalo, F.; Ciccone, R.; Donsante, S.; Volpe, G.; Manni, S.; Guercio, M.; Pezzella, M.; Iaffaldano, L.; et al. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J. Hematol. Oncol. 2022, 15, 163. [Google Scholar] [CrossRef]
  240. Peinert, S.; Prince, H.M.; Guru, P.M.; Kershaw, M.H.; Smyth, M.J.; Trapani, J.A.; Gambell, P.; Harrison, S.; Scott, A.M.; Smyth, F.E.; et al. Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther. 2010, 17, 678–686. [Google Scholar] [CrossRef]
  241. Marofi, F.; Rahman, H.S.; Al-Obaidi, Z.M.J.; Jalil, A.T.; Abdelbasset, W.K.; Suksatan, W.; Dorofeev, A.E.; Shomali, N.; Chartrand, M.S.; Pathak, Y.; et al. Novel CAR T therapy is a ray of hope in the treatment of seriously ill AML patients. Stem Cell Res. Ther. 2021, 12, 465. [Google Scholar] [CrossRef] [PubMed]
  242. Liu, B.; Song, Y.; Liu, D. Clinical trials of CAR-T cells in China. J. Hematol. Oncol. 2017, 10, 166. [Google Scholar] [CrossRef]
  243. Smith, L.; Di Stasi, A. Safety switches used for cellular therapies. In Gene and Cellular Immunotherapy for Cancer; Springer: Berlin/Heidelberg, Germany, 2022; pp. 1–15. [Google Scholar] [CrossRef]
  244. Gargett, T.; Brown, M.P. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front. Pharmacol. 2014, 5, 235. [Google Scholar] [CrossRef]
Figure 1. Targets in multiple myeloma and stages of immunotherapeutic development. This schematic presents targets investigated in CAR-T and CAR-NK cell therapies for MM, categorized by their clinical development stage. Green indicates validated membrane-bound targets with approved CAR-T products (e.g., BCMA). Orange corresponds to targets in Phase I–II trials; light orange to those in Phase I; light pink to targets at the preclinical/Phase I interface; and pink to preclinical-stage targets. The color of each target represents the most advanced development stage achieved for either CAR-T or CAR-NK platforms. Circular markers with “T” or “NK” next to each target indicate whether the target is being explored in CAR-T cells, CAR-NK cells, or both (dual-labeled if applicable), based on available clinical or preclinical evidence. All targets are membrane-bound surface proteins suitable for direct CAR recognition, except APRIL, which is labeled with a “CAR” circle to indicate that it is not a target, but a ligand incorporated into CAR-T constructs as a binding domain, enabling simultaneous of BCMA and TACI. NKG2D ligands (e.g., MICA, MICB, ULBP family) are stress-induced molecules transiently expressed on the tumor cell surface and are targeted using CAR constructs that incorporate the native NKG2D receptor. This figure illustrates the diversity of immunotherapeutic targets in MM and highlights the rationale for multi-target strategies to overcome tumor heterogeneity and antigen escape. Figure created in https://BioRender.com (accessed on 15 June 2025).
Figure 1. Targets in multiple myeloma and stages of immunotherapeutic development. This schematic presents targets investigated in CAR-T and CAR-NK cell therapies for MM, categorized by their clinical development stage. Green indicates validated membrane-bound targets with approved CAR-T products (e.g., BCMA). Orange corresponds to targets in Phase I–II trials; light orange to those in Phase I; light pink to targets at the preclinical/Phase I interface; and pink to preclinical-stage targets. The color of each target represents the most advanced development stage achieved for either CAR-T or CAR-NK platforms. Circular markers with “T” or “NK” next to each target indicate whether the target is being explored in CAR-T cells, CAR-NK cells, or both (dual-labeled if applicable), based on available clinical or preclinical evidence. All targets are membrane-bound surface proteins suitable for direct CAR recognition, except APRIL, which is labeled with a “CAR” circle to indicate that it is not a target, but a ligand incorporated into CAR-T constructs as a binding domain, enabling simultaneous of BCMA and TACI. NKG2D ligands (e.g., MICA, MICB, ULBP family) are stress-induced molecules transiently expressed on the tumor cell surface and are targeted using CAR constructs that incorporate the native NKG2D receptor. This figure illustrates the diversity of immunotherapeutic targets in MM and highlights the rationale for multi-target strategies to overcome tumor heterogeneity and antigen escape. Figure created in https://BioRender.com (accessed on 15 June 2025).
Ijms 26 06051 g001
Table 1. Parameters of tumor targets for CAR therapy.
Table 1. Parameters of tumor targets for CAR therapy.
TargetExpressionExpression in Normal TissuesRisk of Off-Tumor ToxicityDevelopment StageClinical Trial ID (If Available)References
BCMAHigh and stablePlasma cellsLowRegistration (ide-cel, cilta-cel)NCT03361748 and NCT04162210[80,81]
GPRC5DHighSquamous epitheliumLowPhase I/IINCT04555551[82]
FcRH5Moderately highFewLowCase ReportChiCTR2000041025 *[83]
CD38HighImmune and epith. cellsAveragePhase INCT04351022[84]
SLAMF7ModerateNK, DC, and T cellsAveragePhase INCT03958656[85]
CD138HighEpitheliumAveragePhase INCT04430530[86]
CD70ModerateActiv. immune cellsAveragePhase INCT02830724[87]
NKG2D ligandsHeterogenousMinimalLowPhase INCT04623944[88]
CD19Residual (subclones)B cellsAveragePreclinical/Phase INCT02135406[89]
TACIVariableB cellsLowPreclinical
APRILNo data availableNo data availableNo data availablePreclinical
TNFR2ModerateTregs and stromaAverage/highPreclinical
CD44v6VariableKeratinocytesHighPreclinical
Alternative targets for CAR cell therapy in multiple myeloma
CD5670% of casesNK cellsAveragePreclinical
Integrin β7PrevalentlyMinimalLowPreclinical
CD123LowPlasm. DCAveragePreclinical/Phase I (AML)NCT04318678, NCT02159495, NCT02623582, and NCT04599543 (only AML, not MM)[90,91,92,93]
Lewis-Y≈50%LowAveragePhase INCT01716364[94]
*—www.chictr.org.cn as ChiCTR2000041025.
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

Bezborodova, O.A.; Trunova, G.V.; Nemtsova, E.R.; Khokhlova, V.A.; Venediktova, J.B.; Morozova, N.B.; Vorontsova, M.S.; Plyutinskaya, A.D.; Zharova, E.P.; Shegai, P.V.; et al. Targets for CAR Therapy in Multiple Myeloma. Int. J. Mol. Sci. 2025, 26, 6051. https://doi.org/10.3390/ijms26136051

AMA Style

Bezborodova OA, Trunova GV, Nemtsova ER, Khokhlova VA, Venediktova JB, Morozova NB, Vorontsova MS, Plyutinskaya AD, Zharova EP, Shegai PV, et al. Targets for CAR Therapy in Multiple Myeloma. International Journal of Molecular Sciences. 2025; 26(13):6051. https://doi.org/10.3390/ijms26136051

Chicago/Turabian Style

Bezborodova, Olga A., Galina V. Trunova, Elena R. Nemtsova, Varvara A. Khokhlova, Julia B. Venediktova, Natalia B. Morozova, Maria S. Vorontsova, Anna D. Plyutinskaya, Elena P. Zharova, Peter V. Shegai, and et al. 2025. "Targets for CAR Therapy in Multiple Myeloma" International Journal of Molecular Sciences 26, no. 13: 6051. https://doi.org/10.3390/ijms26136051

APA Style

Bezborodova, O. A., Trunova, G. V., Nemtsova, E. R., Khokhlova, V. A., Venediktova, J. B., Morozova, N. B., Vorontsova, M. S., Plyutinskaya, A. D., Zharova, E. P., Shegai, P. V., & Kaprin, A. D. (2025). Targets for CAR Therapy in Multiple Myeloma. International Journal of Molecular Sciences, 26(13), 6051. https://doi.org/10.3390/ijms26136051

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