Next Article in Journal
CT Imaging Patterns in Major Histological Types of Lung Cancer
Previous Article in Journal
Morphology and Function of Red Blood Cells in COVID-19 Patients: Current Overview 2023
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Immune Cells and Immunotherapy in Multiple Myeloma

by
Vijay Radhakrishnan
1,
Upendarrao Golla
2 and
Avinash Kundadka Kudva
3,4,*
1
Department of Surgery, Ellis Fischel Cancer Center, Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA
2
Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
3
Department of Biochemistry, Mangalore University, Mangalagangothri, Mangaluru 574199, India
4
Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
*
Author to whom correspondence should be addressed.
Life 2024, 14(4), 461; https://doi.org/10.3390/life14040461
Submission received: 26 February 2024 / Revised: 28 March 2024 / Accepted: 30 March 2024 / Published: 1 April 2024
(This article belongs to the Section Medical Research)

Abstract

:
The clinical signs of multiple myeloma, a plasma cell (PC) dyscrasia, include bone loss, renal damage, and paraproteinemia. It can be defined as the uncontrolled growth of malignant PCs within the bone marrow. The distinctive bone marrow milieu that regulates the progression of myeloma disease involves interactions between plasma and stromal cells, and myeloid and lymphoid cells. These cells affect the immune system independently or because of a complicated web of interconnections, which promotes disease development and immune evasion. Due to the importance of these factors in the onset of disease, various therapeutic strategies have been created that either target or improve the immunological processes that influence disease progression. The immune system has a role in the mechanism of action of multiple myeloma treatments. The main contributions of immune cells to the bone marrow microenvironment, as well as how they interact and how immune regulation might lead to therapeutic effects, are covered in this study.

1. Introduction

Multiple myeloma (MM) is described by clonally expanding plasma cells within the bone marrow (BM), monoclonal proteins detected in blood or urine, and end-organ damage [1,2]. MM is a cancer that is classified as a clonal B-cell malignancy and is currently incurable. The wide array of immunodeficiencies originating with this plasma cell dyscrasia is an intriguing aspect of the condition [3,4]. As per Cancer Stat Facts published by NIH, about 170,405 myeloma patients were reported from 2017 to 2019, amounting to 1.8% of all new cancer cases in the United States. The estimated new myeloma cases in 2023 are 35,730 (19,860 in men and 15,870 in women), while the number of fatal cases is 12,590 (7000 in males and 5590 in women) [5].
Plasma cells with malignant properties accumulate in bone marrow, causing multiple myeloma. The bone marrow is the soft matter inside bones where blood cells are made. Cancer cells obstruct the growth of healthy blood cells in the bone marrow. Cancer cells produce dysfunctional proteins rather than beneficial antibodies, leading to complications of multiple myeloma.
Bone marrow is thought to be a primary hematopoietic organ. The production and maturation of B lymphocytes take place in the bone marrow. The bone marrow contains the most long-lived plasma cells that can produce antigen-specific antibodies [6]. As a result, bone marrow supports humoral immune reactions. However, a growing body of research shows that immune cells like regulatory T cells, conventional T cells, B cells, dendritic cells, natural killer T (NKT) cells, neutrophils, myeloid-derived suppressor cells, and mesenchymal stem cells are actively functioning and moving around in the bone marrow. In addition, a few human cancers have a preset metastatic site in the bone marrow [7].
In recent years, new therapeutic options have been developed, and our understanding of the biology of the disease has improved [8,9,10]. Patients with myeloma increasingly receive immunomodulatory medications alone or in combination with routine treatment [11,12]. Additionally, adoptive T-cell treatments and immunotherapeutic approaches provide fascinating new research frontiers in myeloma treatment.

2. Role of Immune Cells in Multiple Myeloma (MM)

The immune system detects and destroys pathogens and foreign objects. An essential phase in developing myeloma is the evasion and inhibition of antitumor immunity. MM cells reproduce and proliferate almost exclusively in the BM niche, emphasizing how vital the microenvironment is for promoting cancer progression [13,14,15]. The BM microenvironment is highly vascularized and is made up of hematopoietic cells such as hematopoietic stem cells (HSCs), T and B lymphocytes, myeloid and natural killer (NK) cells, and dendritic cells [16]. The tumor microenvironment (TME) is a highly complex, constantly evolving phenomenon that facilitates bidirectional, mutually advantageous communication between malignant PCs and the BM milieu. TME acts as a protective niche, promoting tumor growth, increasing treatment resistance, and impairing immune surveillance. In MM, complex crosstalk between hematopoietic stem cells, myeloid cells, T and B lymphocytes, NK cells, erythrocytes, osteoclasts, (non-hematopoietic) osteoblasts, stromal cells, e.g., fibroblasts, endothelial cells, and acellular components, e.g., extracellular matrix and cytokines, growth factors, and chemokines produced by cellular components, plays a vital role in tumor progression and immune resistance [17,18]. Hypoxia alters TME and drives many forms of tumor invasion [19]. Hypoxic TMEs cause tumors to become more aggressive and metastatic [20]. Hypoxia-related parameters show predictive potential across malignancies [21]. The hypoxic environment changes the expression of genes that regulate metabolism and other functions. Furthermore, hypoxia signaling interacts with other biological pathways to alter cancer cell malignant behaviors and is intimately related to cancer cell proliferation, migration, invasion, and angiogenesis and affects cancer therapy [22].

2.1. T Cells

MM patients showed fewer CD4+ T cells in peripheral blood, connected to a reduced CD4/CD8 T-cell ratio compared to control healthy individuals [23,24]. In many studies, increased CD4/CD8 T-cell ratio was linked to CD8+ T cells with an activated cytotoxic phenotype (CD8+CD57+CD28−perforin+), but it was also linked to the growth of CD4+ T cells with an effector phenotype (CD28−CD4+). Favorable prognostic indicators have been linked to the surge in T-cell counts in MM patients. The immune system may be attempting to control the proliferation of plasma cells (PCs) utilizing these increased T-cell populations [25,26].
Perez-Andres et al. demonstrated a significantly higher rate of T-cell infiltration of the bone marrow in all patients with MM and plasma cell leukemia. Conversely, less CD4+CD8− and CD4−CD8− T cells were seen. Likewise, the cytotoxic/effector T-cell subsets of CD4+CD28− and CD8+CD28− were also diminished, and in most of the patient groups, TCR expression was elevated in both CD4+ and CD8+ BM T cells [27].

2.2. Helper T Cells

In adaptive immune responses, helper T cells are essential, and the unbalanced polarization of CD4 T-cell responses may significantly impact the progression of multiple myeloma. Though the specifics of the alterations vary, multiple investigations identify a dysregulated cytokine network in MM. Alpana Sharma et al. demonstrated changes in circulatory concentrations of cytokines produced from Th1 and Th2 cells during multiple myeloma [28]. In their study, they compared fifty healthy controls and sixty-two MM patients. They demonstrated that, in terms of serum concentrations of Th1 cytokines, IFN-γ was significantly decreased, and IL-2 was non-significantly higher, whereas IL-4 and IL-10 were considerably enhanced in terms of Th2 cytokines [28]. Th17 T helper cells are derived from naive CD4+ T cells and are implicated in the tumor immunological milieu [29,30]. Numerous investigations have demonstrated that, in comparison to healthy donors, MM patients have considerably higher levels of Th17 cells in PBMCs and serum concentrations of Th17-associated cytokines, such as IL-1b, IL-6, IL-17, IL-21, IL-22, and IL-23 [31,32,33]. Noonan et al. showed that the Th17 T-cell phenotype could be a critical predictor of lytic bone damage in multiple myeloma [34]. MM patients had higher levels of IL-17 than controls, possibly due to elevated levels of IL-6 in their bone marrow, which stimulates the generation of Th17 cells from CD4 naïve cells (as described in previous studies).

2.3. Regulatory T Cells (Tregs)

A vital subgroup of CD4 T-cells known as regulatory T-cells (Tregs) are typified by the CD4 + CD25 + Forkhead box P3+ (FoxP3+) phenotype and are capable of modulating peripheral tolerance as well as responses to foreign and tumor antigens. There is controversy over the role and quantity of Tregs in MM and their relationship to survival metrics, even though Tregs are raised in many forms of malignancy, including hematological malignancies. Treg-mediated inhibition of antitumor immunity has led to an increased frequency of Tregs, widely regarded as a sign of poor prognosis [35,36]. The peripheral blood of MM patients has been found to have a higher frequency of functional Tregs reported by multiple groups [37,38]. In peripheral blood from MM patients, Beyer et al. found that Tregs have a strong inhibitory effect and verified that the patients express higher levels of TGF-β and IL-10 than healthy participants or healthy donors [39].
Higher levels of Treg-associated markers, including CTLA-4, glucocorticoid-induced tumor necrosis factor receptor (GITR), and OX40, have been seen in the peripheral blood of MM patients, which may improve their suppressive function [39].
Tregs are separated into two subpopulations with distinct developmental origins: naïve Tregs (nTregs) and induced Tregs (iTregs). nTregs are progenitor cells in the bone marrow that commit to and mature in the thymus [40]. iTregs are negative selection cells created in the periphery from naïve T-cells. They are classified into type 1 regulatory T-cells (Tr1), which are induced by IL-10 [41,42] and T helper 3 (Th3) cells, which gain regulatory qualities via TGF-β [43,44].
Other lymphocyte subsets with regulatory qualities, such as gamma delta (γδ) T-cells, CD8 + T-cells, double negative cells, and regulatory-B-cells (Bregs), may not necessarily express CD4. Gamma delta T-cells are a small subset of T-cells distinguished by their γδ-T-cell receptors (TCR). They are primarily found in tissues and tumor sites where they exert suppressive activity towards naïve and effector T-cell responses. They also impede the proliferation and function of dendritic cells (DCs) [35]. Andrea Knight et al. found that γδ-T cells have potent cytotoxicity against primary multiple myeloma cells, making them a promising candidate for tumor immunotherapy [45].

2.4. B Cells

The proliferation of mature plasma cells in the bone marrow is a characteristic of MM, a B-cell malignancy. Circulating clonotypic CD19+ cells have been found in several investigations, indicating a development step before the PC. Although it varies greatly, circulating clonotypic CD19+ cells are usually modest. Though a small fraction of circulating clonotypic cells within the CD19 compartment is resistant to even high-dose chemotherapy, these cells react well to induction therapy [46].
B cells are traditionally recognized to positively influence immunological responses and inflammation through antibody production and enhance T-cell activation and proliferation via antigen presentation [47]. However, recent investigations in mice and humans have shown well-founded evidence of separate subsets of immunoregulatory B cells (Bregs/B10 cells) [48,49]. Bregs can maintain immune tolerance, suppress pathological autoimmune and inflammatory immune responses, and suppress responses during cancer immune surveillance by releasing anti-inflammatory mediators like in IL-10 [50,51]. Zhang et al. demonstrated that bone marrow-derived Breg cells inhibit NK cell-mediated ADCC against multiple myeloma cells in multiple myeloma patients by producing IL-10 [52].
B1 cells are a subset of B-cell lymphocytes participating in the humoral immune response [53]. B1a (natural antibody-producing cells) have been demonstrated to contribute to autoimmune disease by autoantibody generation [54,55], antigen presentation and activation of CD4 T cells [56], and cytokine production [57]. Some recent investigations have shown that B1a cells are considerably reduced in MM patients compared to healthy donors in both peripheral blood and bone marrow. The authors also assessed the impact of an immunomodulatory drug (lenalidomide) on B-cell subsets in MM. They analyzed B-cell subsets in PBMC from non-treated and lenalidomide-treated MM patients. They discovered that the lenalidomide-treated group showed significant improvement in B-cell subsets (increased B2 and decreased B1 cells) even before clinical response [58,59].

2.5. Macrophages

The myeloma microenvironment is rich in macrophages. Around 10% of the bone marrow (BM) of patients with multiple sclerosis (MM) is composed primarily of tumor-associated macrophages (TAMs) [60]. TAMs are derived from circulating monocytes that are drawn to the tumor site by cytokines such as VEGF, colony-stimulating factor (CSF)-1, chemokines, and CXC motif chemokine ligand (CXCL)-12 [61,62]. Common surface indicators for identifying macrophages are CD163 and CD206 [63]. Early research by Zheng et al. revealed that MM patients’ bone marrow had a higher concentration of CD68+ macrophages than healthy controls. By preventing caspase-3 and poly-ADP ribose polymerase (PARP) from being activated and cleaved and by preserving Bcl-xL levels in vitro, these macrophages promoted the growth and inhibited apoptosis of myeloma cells in the presence of treatment [60]. Furthermore, it was discovered that macrophages and mesenchymal stromal cells (MSCs) together promote the survival and proliferation of MM cells by producing IL-6 and IL-10 [64].

2.6. Myeloid-Derived Suppressor Cells (MDSCs)

MDSCs are a heterogeneous group of immature myeloid cells produced in the bone marrow [65]. In peripheral blood and bone marrow, a few publications described MDSCs in MM as CD33+ and CD11b+, low or HLA-DR low/-, and further subdivided into CD14 (monocytic MDSc) and CD15 (granulocytic MDSc) [65,66]. On the other hand, CD11b+ and Gr1+ expressions are typically used to identify murine MDSCs [67]. Generating reactive oxygen species (ROS) is necessary for MDSC-dependent immunosuppression. Several studies have reported that MDSCs derived from cancer patients and tumor-bearing mice generate enormous amounts of ROS [68,69], decreasing CD3ζ expression and antigen-specific T-cell proliferation [70]. Purified MDSCs from MM patients produced more Tregs than MDSCs from age-matched controls, according to research by Favaloro J et al. [66]. Various investigations reveal that MDSCs stimulate the clonal growth of antigen-specific natural T-regs and trigger the transformation of naïve T helper cells into inducible T-regs. This process relies on the interaction between CD40 and CD40L and the release of cytokines like TGF-beta, IL-10, and IFN-gamma [71,72]. The role of ROS molecules in the interaction of MDSCs and Tregs is unclear. The induction of Treg cells by macrophages involves ROS production; therefore, ROS deficiency might lead to reduced Treg induction and aggravate T-cell suppression [73]. Wang et al. demonstrated that bortezomib combined with dexamethasone resulted in a progressive decrease in the number of MDSCs. When MM cells and PBMCs were cocultured in the presence of bortezomib, M-MDSC levels were significantly reduced compared to MM cells and PBMCs alone [74]. Another study by Görgün et al. showed that exposure to bortezomib and lenalidomide in vitro did not affect the total quantity or immune suppressive capacity of MDSCs [75]. MDSCs, especially G-MDSCs, may interact with MM cells bidirectionally, implying that treatments targeting M-MDSCs may improve therapeutic outcomes in MM patients.

2.7. Natural Killer (NK) and Natural Killer T Cells (NKT Cells)

An integral part of the innate immune system, NK cells are cytotoxic lymphocytes linked to antitumor immunity [76]. Hematopoietic stem cells give rise to natural killer (NK) cells, which proliferate and acquire effector capabilities such as cytokine production and lysis capacity when exposed to IL-2 and IL-15 [77]. NKT cells constitute a conserved T-cell sub-lineage with unique properties, including reactivity for a synthetic glycolipid presented by CD1d and common natural killer cell markers such as NK1.1 in mice or CD161 in humans, expressing CD122 (IL-2Rb), Ly49 family receptors, and T-cell receptor [78,79]. The decrease in NK and NKT cells’ ability to secrete IFN-gamma is linked to the development of clinical myeloma, as revealed by Dhodapkar MV et al. [80,81]. Several studies showed that premalignant and early myeloma tumor cells had significantly elevated CD1d levels [82]. As the disease progressed, CD1d expression decreased and finally vanished completely, which was detrimental to myeloma cell survival [82].

2.8. Dendritic Cells (DCs)

DCs are significant antigen-presenting cells that can present autoantigens or foreign antigens [83,84]. DCs show expression on their surface, including DC-SIGN, CD1a, CD40, CD54, CD58, CD80, CD83, and CD86. Additionally, they have significant levels of MHC molecule class I and II expressions [85]. Typically, DCs are essential for plasma cell survival and differentiation [86,87]. A study by Brimnes et al. revealed that when MM patients were compared to healthy controls, there was a decrease in the quantity and expression of human leucocyte antigen (HLA) molecules on both myeloid and plasmacytoid DCs [88]. Moreover, MM patients’ CCR5, CCR7, and DEC205 expressions were lower than regular donors. According to Leone et al., DCs are said to accumulate in BM as MM progresses. The study demonstrated that DCs isolated from myeloma patients could engulf apoptotic myeloma cells, present them, and activate CD8+ T lymphocytes specific to tumors. Additionally, DCs inhibited the production of proteasome subunits in myeloma cells [89].
Myeloma pathology is also associated with plasmacytoid DCs (pDCs), the other major component of DCs. The production of normal plasma cells and antibody responses primarily depend on the secretion of IFN-gamma and IL-6 by pDCs [87,90]. MM patients have higher frequencies and quantities of BM pDCs, and studies by Chauhan et al. have demonstrated that pDCs give myeloma cells the ability to develop, survive, chemotaxis, and resist drugs [91]. pDC cells facilitate MM development and medication resistance. In vitro, MM-conditioned-pDCs lose their ability to release IFN-α, indicating a direct interaction between MM cells and pDCs, leading to a tumor-promoting phenotype [92]. Thus, to enhance myeloma development and avoid immune surveillance, MM cells regulate the function of pDCs via the CDH1 pathway. The molecular mechanisms driving pDC acquisition of these traits are unknown, contradicting findings on the amount of DCs in MM [89,91,93,94].

3. Regulation of Immune Cells by Immunotherapy in Multiple Myeloma

Immunotherapeutic agents used against multiple myeloma work in distinct ways. This section discusses some leading strategies and targeted actions that have emerged as promising treatments against MM.

3.1. Immunomodulatory Drugs

Immunomodulatory drugs (IMiDs) modulate the immune response by either stimulating the formation of serum antibodies (immunosuppressives) or inhibiting them (immunomodulators) [95]. IMiDs are a class of medications that exhibit many antimyeloma properties, including immunomodulation, anti-angiogenic, anti-inflammatory, and antiproliferative effects [96,97].
IMiDs mediate antimyeloma action via direct and indirect mechanisms [98]. The proliferation and survival of myeloma cells were not significantly affected by first-generation IMiDs, such as thalidomide. These medications were employed initially as hypotonic sedatives and were later withdrawn because of teratogenic consequences [99]. However, because of their anti-angiogenic and immunomodulatory qualities, they were reinstated as a therapy option for multiple myeloma. Singhal et al. treated eighty-four patients with refractory myeloma, in which seventy-six patients relapsed after a high dose of chemotherapy received oral thalidomide as a single agent for a median of 80 days. Authors observed durable responses in some patients with multiple myeloma, including those who relapse after high-dose chemotherapy [100]. Four distinct phase III trials compared the use of thalidomide in conjunction with melphalan or prednisone in transplant-eligible patients. One of the experiments included thalidomide maintenance until relapse [101]. In MM cells, second-generation IMiDs can directly cause cell cycle arrest and apoptosis [102,103]. IMiDs are the cornerstone of myeloma treatment, and in clinical trials, they typically serve as the gold standard to which other medications are added. In phase II research, Niesvizky et al. demonstrated that adding clarithromycin to lenalidomide and dexamethasone (BiRD) resulted in a greater overall response rate [104]. In another trial, lenalidomide demonstrated potential activity as a maintenance medication after autologous stem cell transplantation. McCarthy et al. found a median duration to advancement of 46 months versus 27 months [105]. Monoclonal antibodies, steroids, protease inhibitors, and epigenetic regulators are combined with IMiDs [106,107]. Primary medications used to treat MM are listed in Table 1.
IMiDs can interact with various cells, including immune cells, osteoclasts, and bone marrow stromal cells, which interfere with the interaction between the MM cells and the bone marrow milieu, explaining the indirect mechanisms. The interaction between MM cells and the bone marrow microenvironment promotes cell growth, survival, migration, and drug resistance. IMiDs work against MM clone proliferation by acting as anti-angiogenic and anti-inflammatory agents. They also mediate cytokine production and surface adhesion molecule expression on MM and bone marrow stromal cells [130,131]. According to various reports, IMiDs suppress the expression of bFGF and VEGF, prevent the synthesis of TNF-α, IL-1, IL-6, and IL-12, delay the development of osteoclasts, and alter the activity of some immune system cells [132,133].
IMiDs differ from other antimyeloma medications solely by their immunomodulatory effect. Thalidomide is less effective than second-generation IMiDs in promoting CD4+ and CD8+ T-cell co-stimulation (Figure 1). Tregs are also crucial for maintaining the body’s immunological tolerance and share many molecular signaling pathways with conventional T cells, especially cytotoxic T cells, which are principal mediators of tumor immunity [132,134,135]. The potential synergistic effect of this immunomodulatory function with emerging immunotherapeutic drugs like checkpoint inhibitors and monoclonal antibodies makes it intriguing.
NK and NKT cells are enhanced by IMiDs [136]. They promote NK cell proliferation in MM patients responding to therapy and enhance NK cell-mediated cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC) induced by triggering IL-2 production from T cells [137].

3.2. Immune Checkpoint Inhibitors (ICIs)

The survival rates of numerous solid tumors have significantly increased with immune checkpoint inhibitors [138]. As a hematologic malignancy, MM has a clear indication that the tumor’s pathophysiology was influenced by a compromised immune system [139]. ICIs cause cancer cells to undergo apoptosis, stop their proliferation, and block receptors that inactivate lymphocytes [140]. Programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and programmed death-ligand 1 (PD-L1) are different types of ICIs. T cells, a subset of white blood cells directly supporting the body’s immune system in battling illness, have PD-1 on their surface. Blocking PD-1 improves the immune system’s ability to eradicate the disease by preventing the body from eliminating cancer cells [141,142]. PD-L1 on MM cells inhibits the activation potential and triggers T-cell activation [142]. CTLA-4 is a receptor present on the surface of T cells. CTLA-4 expression was seen upon activation of T cells, including memory and regulatory T-cells [143]. It has been demonstrated that patients with MM have higher levels of CTLA-4 and PD-L1 expression in their bone marrow microenvironment [144]. Additional checkpoint inhibitors that target T-cell immunoglobulin 3 (TIM3), T-cell immunoreceptors with immunoglobulin and ITIM domains (TIGIT), and lymphocyte activation gene-3 (LAG3) are under investigation [145].
A phase I multicenter, multi-cohort study in patients with RRMM that combined the PD-1 inhibitor pembrolizumab, lenalidomide, and low-dose dexamethasone reported that 62 patients had a 44% ORR. Another study confirmed the efficacy of bonadomide and lenalidomide in enhancing ICIs in myeloma cells. In contrast, a combination of dexamethasone, lenalidomide, and the PD-L1 antibody pembrolizumab was effective in patients with lenalidomide-refractory myeloma, with an objective response rate of 76% [146].

3.3. Chimeric Antigen Receptor (CAR)-T Cell Therapy

Despite a broad range of approved treatments, relapsed/refractory multiple myeloma (RRMM) continues to pose a serious clinical challenge. T-cells engineered with chimeric antigen receptors (CARs) have revolutionized the treatment of patients with hematologic malignancies. CAR-T cells targeting CD-19 B-cell lymphoma cells have demonstrated significant efficacy in treating acute lymphoblastic leukemia and lymphoma [147]. The treatment of MM with CAR T cell therapy has shown considerable promise [148]. The B-cell maturation antigen is the most often tested target for MM. Host cells modified to express an antigen recognition domain capable of binding a specific target for tumors are called CAR-T cells [149,150]. CAR-T receptors are manufactured transmembrane proteins containing antigen recognition domains linked by a linker, followed by costimulatory and T-cell activation domains [151].

3.3.1. B-Cell Maturation Antigen (BCMA) CAR-T

The highly selective expression of B-cell maturation antigen (BCMA) in malignant plasma cells makes it a novel target for MM treatment. BCMA is also known as tumor necrosis factor receptor superfamily member 17 (TNFRSF17) or CD269. A crucial element of plasma cell homeostasis, this transmembrane protein controls B-cell development and differentiation into plasma cells [152]. BCMA’s correlation with plasma cell survival satisfies the functional dependency condition for an ideal target [153]. The higher levels of its ligands facilitate the proliferative and anti-apoptotic behavior of MM, B-cell activating factor (BAFF), and a proliferation-inducing ligand (APRIL) [154]. In the first-in-human BCMA CAR-T study, researchers at the National Cancer Institute (NCI) employed CD28 for the costimulatory domain, retrovirus vector for transduction, and murine scFv for antigen recognition [155]. Their remarkable 81% objective response rate (ORR) was reported. Subsequently, the outcomes of other BCMA CAR-T clinical trials were released. Selected clinical studies of BCMA CAR-T are summarized in Table 2.

3.3.2. Non-BCMA CAR-T

In patients with RRMM, CAR-T cell treatments with BCMA have produced extremely encouraging outcomes. Studies conducted on patients receiving BCMA CAR-T treatments over an extended period showed no relapses [166]. CAR-T cell treatments targeting non-BCMA antigens have demonstrated efficacious outcomes in MM patients [167]. When combined with BCMA, new targets exhibit extremely encouraging results. Non-BCMA targets include CD138 (syndecan-1), CD38, NY-ESO-1, NKG2D ligands, SLAMF7/CS1, CD56, Lewis Y, and GPRC5D.
Cluster of Differentiation 19 (CD19) is a transmembrane protein encoded by the CD19 gene. All human B-cell lineages express CD19. The myeloma plasma cell surface exhibits a low expression of CD19; CD19+ cells may be a subgroup of stem cells in patients with myeloma. Several studies demonstrated that CD19 CART cells with anti-BCMA therapies showed promising efficacy in treating MM [168]. Recently, G-Protein Coupled Receptor 5D (GPRC5D) has been found as another possible target for MM because this antigen is expressed at high levels on malignant MM cells regardless of BCMA distribution and only at low levels on B cells, healthy plasma cells, and hair follicles [169]. MM cells exhibit high expression of CD138, commonly known as syndecan-1. According to a study, CD138-directed CART cells can eradicate primary myeloma cells in vivo and in vitro [170]. The New York Esophageal Squamous Cell Carcinoma 1 (NY-ESO-1) testis gene is highly immunogenic and often expressed in many cancer types [171]. TCR-engineered T cells specific to NY-ESO-1 have produced positive therapeutic outcomes in MM patients [172].
The signaling lymphocytic activation molecule (SLAM) receptor family includes the SLAMF7 (CD319, CS-1) receptor. SLAMF7 can influence various immune cell-specific functions in several cell types [173]. The first FDA-approved antibody for the treatment of MM was elotuzumab, a humanized monoclonal antibody targeted to SLAMF7. Emotruzaba, in conjunction with lenalidomide or pomalidomide, has demonstrated therapeutic effectiveness in patients with relapsed multiple myeloma [174].
NKG2D (natural-killer group 2, member D) receptor plays a vital role in protecting the host from infections and cancer. NKG2D is one of the best-characterized activating immune receptors associated with tumor immunosurveillance [175,176]. Both primary and metastatic cancer cells frequently upregulate stress-induced ligands closely related to MHC class I [177]. NKG2D CAR T-cell therapy showed a therapeutic effect on MM, lymphoma, and ovarian cancers in vivo [178]. Table 3 summarizes the selected non-BCMA-targeted CAR-T clinical studies.

3.3.3. CAR-T Therapy Limitations and Toxicities

Despite its current success rate, CAR-T treatment is associated with substantial toxicities. The most prevalent adverse effects associated with CAR-T treatment, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), continue to be hurdles to its usage. CRS is the most common type of toxicity associated with CAR-T treatment. Symptoms typically appear within the first week following CAR-T cell injection and last 7 to 8 days [188]. CRS is considered a reversible consequence of CAR-T treatment, occurring in 42% to 100% of patients. It manifests with varied intensity, with 0 to 46% of patients presenting severe CRS symptoms and 0 to 9.1% of cases proceeding to death [189]. ICANS, like CRS, is a transient issue that occurs in the majority of patients following CAR-T infusion. It is less common and more typically delayed than CRS, with an incidence ranging from 3 to 64% and 0 to 54% of cases presenting indications of severe disease [190]. It is a clinical and neuropsychiatric illness manifestation that develops between days to 2–3 weeks of CAR-T treatment administration [191]. Toxicology prevention and management strategies have been presented from a variety of perspectives. Optimization of dosing schemes is a realistic clinical strategy, while laboratory strategies involve creating less toxic CAR structures, which could considerably increase the safety of CAR T-cell treatment. However, further research is necessary before these treatments can be extensively used in clinical settings.

4. Limitations of Immunotherapy

Developing inherent or acquired resistance is a significant hurdle in cancer immunotherapy research. Cancer cells have a variety of methods to avoid immunosurveillance, which causes resistance to immunotherapy, such as checkpoint inhibitors, CAR-T cell treatment, and recombinant cytokines [192]. Immunotherapies are commonly limited by Immune-Related Adverse Events (irAEs), which include impervious actuation and an incendiary reaction against the host’s healthy tissues. Although irAEs aim to forecast, evaluate, and cure, the ideal outcome is resistant actuation against host development [193,194].
Despite solid response rates in numerous BCMA-targeted CAR T-cell treatments, response durability remains a clinical hurdle in the treatment of MM. A significant number of patients develop recurrence [195]. Other challenges include the inability to predict the efficacy of the therapy and the patient’s response, the need for additional biomarkers, the improvement in immunity in opposition to malignant immunotherapies, the lack of up-to-date medical assessment plans to determine sufficiency, and high therapy costs.

5. Future Perspectives in the Field of Immunotherapy

Checkpoint inhibitors, antibody-based medicines, adoptive cell therapy, BCMA, and non-BCMA CAR-T treatments are all promising advances in treating multiple myeloma [196,197]. Ongoing clinical trials are testing bispecific T cell antibodies in conjunction with other therapies, such as IMiDs and PIs, with the potential for further anti-myeloma action. Furthermore, current research investigating the combination of BCMA targeting diverse antigens on plasma cells is particularly intriguing, hinting at the prospect of pure immunotherapy for MM, eliminating the toxicity of traditional therapy. To reduce the risk of recurrence in multiple myeloma after CAR T-cell therapy, researchers are concentrating on new antigens that can be targeted alone or in combination with BCMA [198]. Furthermore, pharmacological medications can increase the density of target antigens on MM cells, thereby enhancing patient outcomes. Exploring the impact of specific genomic and transcriptomic markers, such as T-cells and RNA editing signatures, on surface target expression may reveal new paths for immune targeting in MM patients. Ongoing research aims to discover techniques to increase target antigen expression, improve immunotherapy durability, and overcome immunosuppressive obstacles in the tumor microenvironment. These improvements show promise for the future of multiple myeloma therapy.

6. Summary

This study investigated the intricate relationship between immune cells and cancer and their function in halting the formation of tumors and the disease. Understanding the potential of immune-based therapy and cancer therapies is facilitated by this review. Identification of antigens associated with myeloma and strategies to reverse tumor-mediated immune suppression are critical components of effective immunotherapy. Regression of the disease seems to be highly induced by antibody therapy, either by itself or in combination with other anti-MM drugs. While checkpoint blocking has been shown to have potential as a cancer treatment, combined therapy may be necessary for checkpoint blocking to be effective in multiple myeloma. The immune system has developed into a potent tool because of its capacity to recognize cancer cells, capture tumor heterogeneity, and supply memory to prevent a recurrence. For MM patients, the long-term prognosis is expected to be significantly improved by developing cell therapies in combination with immunomodulatory drugs, immune checkpoint inhibitors, and CARTs.

Author Contributions

V.R. conceived the structure of the initial draft of this review and investigated key resources for the review. V.R., A.K.K. and U.G. wrote, proofread, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge respective affiliated universities/institutions for their support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Munshi, N.C.; Avet-Loiseau, H.; Rawstron, A.C.; Owen, R.G.; Child, J.A.; Thakurta, A.; Sherrington, P.; Samur, M.K.; Georgieva, A.; Anderson, K.C.; et al. Association of Minimal Residual Disease with Superior Survival Outcomes in Patients with Multiple Myeloma: A Meta-analysis. JAMA Oncol. 2017, 3, 28–35. [Google Scholar] [CrossRef] [PubMed]
  2. Palumbo, A.; Anderson, K. Multiple myeloma. N. Engl. J. Med. 2011, 364, 1046–1060. [Google Scholar] [CrossRef] [PubMed]
  3. Zaknoen, S.L.; Kay, N.E. Immunoregulatory cell dysfunction in chronic B-cell leukemias. Blood Rev. 1990, 4, 165–174. [Google Scholar] [CrossRef] [PubMed]
  4. Barlogie, B.; Epstein, J.; Selvanayagam, P.; Alexanian, R. Plasma cell myeloma—New biological insights and advances in therapy. Blood 1989, 73, 865–879. [Google Scholar] [CrossRef] [PubMed]
  5. ACS. Key Statistics about Multiple Myeloma. Available online: https://www.cancer.org/cancer/types/multiple-myeloma.html (accessed on 25 February 2024).
  6. Kopp, H.G.; Avecilla, S.T.; Hooper, A.T.; Rafii, S. The bone marrow vascular niche: Home of HSC differentiation and mobilization. Physiology 2005, 20, 349–356. [Google Scholar] [CrossRef] [PubMed]
  7. Zhao, E.; Xu, H.; Wang, L.; Kryczek, I.; Wu, K.; Hu, Y.; Wang, G.; Zou, W. Bone marrow and the control of immunity. Cell. Mol. Immunol. 2012, 9, 11–19. [Google Scholar] [CrossRef]
  8. Harousseau, J.L. Multiple myeloma in the elderly: When to treat, when to go to transplant. Oncology 2010, 24, 992–998. [Google Scholar] [PubMed]
  9. Kumar, A.; Galeb, S.; Djulbegovic, B. Treatment of patients with multiple myeloma: An overview of systematic reviews. Acta Haematol. 2011, 125, 8–22. [Google Scholar] [CrossRef] [PubMed]
  10. Shah, N.; Lonial, S. Evidence-based mini-review: Treatment options for patients with relapsed/refractory myeloma previously treated with novel agents and high-dose chemotherapy and autologous stem-cell transplantation. Hematol. Am. Soc. Hematol. Educ. Program 2010, 2010, 310–313. [Google Scholar] [CrossRef]
  11. Serafini, P.; Meckel, K.; Kelso, M.; Noonan, K.; Califano, J.; Koch, W.; Dolcetti, L.; Bronte, V.; Borrello, I. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J. Exp. Med. 2006, 203, 2691–2702. [Google Scholar] [CrossRef]
  12. van de Donk, N.W.; Lokhorst, H.M.; Dimopoulos, M.; Cavo, M.; Morgan, G.; Einsele, H.; Kropff, M.; Schey, S.; Avet-Loiseau, H.; Ludwig, H.; et al. Treatment of relapsed and refractory multiple myeloma in the era of novel agents. Cancer Treat. Rev. 2011, 37, 266–283. [Google Scholar] [CrossRef] [PubMed]
  13. Kawano, Y.; Moschetta, M.; Manier, S.; Glavey, S.; Gorgun, G.T.; Roccaro, A.M.; Anderson, K.C.; Ghobrial, I.M. Targeting the bone marrow microenvironment in multiple myeloma. Immunol. Rev. 2015, 263, 160–172. [Google Scholar] [CrossRef] [PubMed]
  14. Leone, P.; Solimando, A.G.; Malerba, E.; Fasano, R.; Buonavoglia, A.; Pappagallo, F.; De Re, V.; Argentiero, A.; Silvestris, N.; Vacca, A.; et al. Actors on the Scene: Immune Cells in the Myeloma Niche. Front. Oncol. 2020, 10, 599098. [Google Scholar] [CrossRef] [PubMed]
  15. Podar, K.; Chauhan, D.; Anderson, K.C. Bone marrow microenvironment and the identification of new targets for myeloma therapy. Leukemia 2009, 23, 10–24. [Google Scholar] [CrossRef] [PubMed]
  16. Cowan, A.J.; Green, D.J.; Kwok, M.; Lee, S.; Coffey, D.G.; Holmberg, L.A.; Tuazon, S.; Gopal, A.K.; Libby, E.N. Diagnosis and Management of Multiple Myeloma: A Review. JAMA 2022, 327, 464–477. [Google Scholar] [CrossRef] [PubMed]
  17. 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] [PubMed]
  18. Manier, S.; Sacco, A.; Leleu, X.; Ghobrial, I.M.; Roccaro, A.M. Bone marrow microenvironment in multiple myeloma progression. J. Biomed. Biotechnol. 2012, 2012, 157496. [Google Scholar] [CrossRef]
  19. Qureshi-Baig, K.; Kuhn, D.; Viry, E.; Pozdeev, V.I.; Schmitz, M.; Rodriguez, F.; Ullmann, P.; Koncina, E.; Nurmik, M.; Frasquilho, S.; et al. Hypoxia-induced autophagy drives colorectal cancer initiation and progression by activating the PRKC/PKC-EZR (ezrin) pathway. Autophagy 2020, 16, 1436–1452. [Google Scholar] [CrossRef] [PubMed]
  20. Semenza, G.L. Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharmacol. Sci. 2012, 33, 207–214. [Google Scholar] [CrossRef]
  21. Zhang, Q.; Huang, R.; Hu, H.; Yu, L.; Tang, Q.; Tao, Y.; Liu, Z.; Li, J.; Wang, G. Integrative Analysis of Hypoxia-Associated Signature in Pan-Cancer. iScience 2020, 23, 101460. [Google Scholar] [CrossRef]
  22. Luo, Z.; Tian, M.; Yang, G.; Tan, Q.; Chen, Y.; Li, G.; Zhang, Q.; Li, Y.; Wan, P.; Wu, J. Hypoxia signaling in human health and diseases: Implications and prospects for therapeutics. Signal Transduct. Target. Ther. 2022, 7, 218. [Google Scholar] [CrossRef] [PubMed]
  23. Mills, K.H.; Cawley, J.C. Abnormal monoclonal antibody-defined helper/suppressor T-cell subpopulations in multiple myeloma: Relationship to treatment and clinical stage. Br. J. Haematol. 1983, 53, 271–275. [Google Scholar] [CrossRef] [PubMed]
  24. Raitakari, M.; Brown, R.D.; Gibson, J.; Joshua, D.E. T cells in myeloma. Hematol. Oncol. 2003, 21, 33–42. [Google Scholar] [CrossRef]
  25. Hayashi, T.; Hideshima, T.; Akiyama, M.; Raje, N.; Richardson, P.; Chauhan, D.; Anderson, K.C. Ex vivo induction of multiple myeloma-specific cytotoxic T lymphocytes. Blood 2003, 102, 1435–1442. [Google Scholar] [CrossRef]
  26. Davies, F.E.; Raje, N.; Hideshima, T.; Lentzsch, S.; Young, G.; Tai, Y.T.; Lin, B.; Podar, K.; Gupta, D.; Chauhan, D.; et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood 2001, 98, 210–216. [Google Scholar] [CrossRef]
  27. Perez-Andres, M.; Almeida, J.; Martin-Ayuso, M.; Moro, M.J.; Martin-Nunez, G.; Galende, J.; Hernandez, J.; Mateo, G.; San Miguel, J.F.; Orfao, A.; et al. Characterization of bone marrow T cells in monoclonal gammopathy of undetermined significance, multiple myeloma, and plasma cell leukemia demonstrates increased infiltration by cytotoxic/Th1 T cells demonstrating a squed TCR-Vbeta repertoire. Cancer 2006, 106, 1296–1305. [Google Scholar] [CrossRef]
  28. Sharma, A.; Khan, R.; Joshi, S.; Kumar, L.; Sharma, M. Dysregulation in T helper 1/T helper 2 cytokine ratios in patients with multiple myeloma. Leuk. Lymphoma 2010, 51, 920–927. [Google Scholar] [CrossRef]
  29. Guery, L.; Hugues, S. Th17 Cell Plasticity and Functions in Cancer Immunity. BioMed Res. Int. 2015, 2015, 314620. [Google Scholar] [CrossRef]
  30. Asadzadeh, Z.; Mohammadi, H.; Safarzadeh, E.; Hemmatzadeh, M.; Mahdian-Shakib, A.; Jadidi-Niaragh, F.; Azizi, G.; Baradaran, B. The paradox of Th17 cell functions in tumor immunity. Cell. Immunol. 2017, 322, 15–25. [Google Scholar] [CrossRef]
  31. Prabhala, R.H.; Pelluru, D.; Fulciniti, M.; Prabhala, H.K.; Nanjappa, P.; Song, W.; Pai, C.; Amin, S.; Tai, Y.T.; Richardson, P.G.; et al. Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood 2010, 115, 5385–5392. [Google Scholar] [CrossRef]
  32. Bryant, C.; Suen, H.; Brown, R.; Yang, S.; Favaloro, J.; Aklilu, E.; Gibson, J.; Ho, P.J.; Iland, H.; Fromm, P.; et al. Long-term survival in multiple myeloma is associated with a distinct immunological profile, which includes proliferative cytotoxic T-cell clones and a favourable Treg/Th17 balance. Blood Cancer J. 2013, 3, e148. [Google Scholar] [CrossRef]
  33. Kara, E.E.; McKenzie, D.R.; Bastow, C.R.; Gregor, C.E.; Fenix, K.A.; Ogunniyi, A.D.; Paton, J.C.; Mack, M.; Pombal, D.R.; Seillet, C.; et al. CCR2 defines in vivo development and homing of IL-23-driven GM-CSF-producing Th17 cells. Nat. Commun. 2015, 6, 8644. [Google Scholar] [CrossRef]
  34. Noonan, K.; Marchionni, L.; Anderson, J.; Pardoll, D.; Roodman, G.D.; Borrello, I. A novel role of IL-17-producing lymphocytes in mediating lytic bone disease in multiple myeloma. Blood 2010, 116, 3554–3563. [Google Scholar] [CrossRef]
  35. Hadjiaggelidou, C.; Katodritou, E. Regulatory T-Cells and Multiple Myeloma: Implications in Tumor Immune Biology and Treatment. J. Clin. Med. 2021, 10, 4588. [Google Scholar] [CrossRef] [PubMed]
  36. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef]
  37. Muthu Raja, K.R.; Rihova, L.; Zahradova, L.; Klincova, M.; Penka, M.; Hajek, R. Increased T regulatory cells are associated with adverse clinical features and predict progression in multiple myeloma. PLoS ONE 2012, 7, e47077. [Google Scholar] [CrossRef]
  38. Feyler, S.; von Lilienfeld-Toal, M.; Jarmin, S.; Marles, L.; Rawstron, A.; Ashcroft, A.J.; Owen, R.G.; Selby, P.J.; Cook, G. CD4+CD25+FoxP3+ regulatory T cells are increased whilst CD3+CD4CD8alphabetaTCR+ Double Negative T cells are decreased in the peripheral blood of patients with multiple myeloma which correlates with disease burden. Br. J. Haematol. 2009, 144, 686–695. [Google Scholar] [CrossRef]
  39. Beyer, M.; Kochanek, M.; Giese, T.; Endl, E.; Weihrauch, M.R.; Knolle, P.A.; Classen, S.; Schultze, J.L. In vivo peripheral expansion of naive CD4+CD25high FoxP3+ regulatory T cells in patients with multiple myeloma. Blood 2006, 107, 3940–3949. [Google Scholar] [CrossRef]
  40. Sakaguchi, S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu. Rev. Immunol. 2004, 22, 531–562. [Google Scholar] [CrossRef]
  41. Groux, H.; O’Garra, A.; Bigler, M.; Rouleau, M.; Antonenko, S.; de Vries, J.E.; Roncarolo, M.G. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 1997, 389, 737–742. [Google Scholar] [CrossRef]
  42. Vieira, P.L.; Christensen, J.R.; Minaee, S.; O’Neill, E.J.; Barrat, F.J.; Boonstra, A.; Barthlott, T.; Stockinger, B.; Wraith, D.C.; O’Garra, A. IL-10-secreting regulatory T cells do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+CD25+ regulatory T cells. J. Immunol. 2004, 172, 5986–5993. [Google Scholar] [CrossRef] [PubMed]
  43. Fehervari, Z.; Sakaguchi, S. Development and function of CD25+CD4+ regulatory T cells. Curr. Opin. Immunol. 2004, 16, 203–208. [Google Scholar] [CrossRef] [PubMed]
  44. Weiner, H.L. Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells. Immunol. Rev. 2001, 182, 207–214. [Google Scholar] [CrossRef] [PubMed]
  45. Knight, A.; Mackinnon, S.; Lowdell, M.W. Human Vdelta1 gamma-delta T cells exert potent specific cytotoxicity against primary multiple myeloma cells. Cytotherapy 2012, 14, 1110–1118. [Google Scholar] [CrossRef]
  46. Bagg, A.; Becker, P.; Bezwoda, W.; van Rensburg, L.; Mendelow, B. Circulating monotypic B-cells in multiple myeloma: Association with lambda paraproteins. Br. J. Haematol. 1989, 72, 167–172. [Google Scholar] [CrossRef]
  47. LeBien, T.W.; Tedder, T.F. B lymphocytes: How they develop and function. Blood 2008, 112, 1570–1580. [Google Scholar] [CrossRef] [PubMed]
  48. Yanaba, K.; Bouaziz, J.D.; Haas, K.M.; Poe, J.C.; Fujimoto, M.; Tedder, T.F. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity 2008, 28, 639–650. [Google Scholar] [CrossRef]
  49. Blair, P.A.; Norena, L.Y.; Flores-Borja, F.; Rawlings, D.J.; Isenberg, D.A.; Ehrenstein, M.R.; Mauri, C. CD19+CD24hiCD38hi B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity 2010, 32, 129–140. [Google Scholar] [CrossRef]
  50. Rosser, E.C.; Mauri, C. Regulatory B cells: Origin, phenotype, and function. Immunity 2015, 42, 607–612. [Google Scholar] [CrossRef]
  51. Khan, A.R.; Hams, E.; Floudas, A.; Sparwasser, T.; Weaver, C.T.; Fallon, P.G. PD-L1hi B cells are critical regulators of humoral immunity. Nat. Commun. 2015, 6, 5997. [Google Scholar] [CrossRef]
  52. Zhang, L.; Tai, Y.T.; Ho, M.; Xing, L.; Chauhan, D.; Gang, A.; Qiu, L.; Anderson, K.C. Regulatory B cell-myeloma cell interaction confers immunosuppression and promotes their survival in the bone marrow milieu. Blood Cancer J. 2017, 7, e547. [Google Scholar] [CrossRef] [PubMed]
  53. Cunningham, A.F.; Flores-Langarica, A.; Bobat, S.; Dominguez Medina, C.C.; Cook, C.N.; Ross, E.A.; Lopez-Macias, C.; Henderson, I.R. B1b cells recognize protective antigens after natural infection and vaccination. Front. Immunol. 2014, 5, 535. [Google Scholar] [CrossRef] [PubMed]
  54. Zhong, X.; Lau, S.; Bai, C.; Degauque, N.; Holodick, N.E.; Steven, S.J.; Tumang, J.; Gao, W.; Rothstein, T.L. A novel subpopulation of B-1 cells is enriched with autoreactivity in normal and lupus-prone mice. Arthritis Rheum. 2009, 60, 3734–3743. [Google Scholar] [CrossRef] [PubMed]
  55. Enghard, P.; Humrich, J.Y.; Chu, V.T.; Grussie, E.; Hiepe, F.; Burmester, G.R.; Radbruch, A.; Berek, C.; Riemekasten, G. Class switching and consecutive loss of dsDNA-reactive B1a B cells from the peritoneal cavity during murine lupus development. Eur. J. Immunol. 2010, 40, 1809–1818. [Google Scholar] [CrossRef] [PubMed]
  56. Zhong, X.; Gao, W.; Degauque, N.; Bai, C.; Lu, Y.; Kenny, J.; Oukka, M.; Strom, T.B.; Rothstein, T.L. Reciprocal generation of Th1/Th17 and T(reg) cells by B1 and B2 B cells. Eur. J. Immunol. 2007, 37, 2400–2404. [Google Scholar] [CrossRef] [PubMed]
  57. Maseda, D.; Candando, K.M.; Smith, S.H.; Kalampokis, I.; Weaver, C.T.; Plevy, S.E.; Poe, J.C.; Tedder, T.F. Peritoneal cavity regulatory B cells (B10 cells) modulate IFN-gamma+CD4+ T cell numbers during colitis development in mice. J. Immunol. 2013, 191, 2780–2795. [Google Scholar] [CrossRef] [PubMed]
  58. Prabhala, R.H.; Talluri, S.; Stekla, M.; Negrroiu, A.; Buonopane, M.; LaBlanc, K.; Heubeck, A.; Fulciniti, M.; Anderson, K.; Munshi, N. Defining Fundamental B Cell-Subset Dysfunction in Myeloma. Blood 2016, 128, 2085. [Google Scholar] [CrossRef]
  59. Plante, M.; Kwon, J.S.; Ferguson, S.; Samouëlian, V.; Ferron, G.; Maulard, A.; de Kroon, C.; Driel, W.V.; Tidy, J.; Marth, C.; et al. An international randomized phase III trial comparing radical hysterectomy and pelvic node dissection (RH) vs simple hysterectomy and pelvic node dissection (SH) in patients with low-risk early-stage cervical cancer (LRESCC): A Gynecologic Cancer Intergroup study led by the Canadian Cancer Trials Group (CCTG CX.5-SHAPE). J. Clin. Oncol. 2023, 41, LBA5511. [Google Scholar] [CrossRef]
  60. Zheng, Y.; Cai, Z.; Wang, S.; Zhang, X.; Qian, J.; Hong, S.; Li, H.; Wang, M.; Yang, J.; Yi, Q. Macrophages are an abundant component of myeloma microenvironment and protect myeloma cells from chemotherapy drug-induced apoptosis. Blood 2009, 114, 3625–3628. [Google Scholar] [CrossRef]
  61. Petty, A.J.; Yang, Y. Tumor-Associated Macrophages in Hematologic Malignancies: New Insights and Targeted Therapies. Cells 2019, 8, 1526. [Google Scholar] [CrossRef]
  62. Ribatti, D.; Moschetta, M.; Vacca, A. Macrophages in multiple myeloma. Immunol. Lett. 2014, 161, 241–244. [Google Scholar] [CrossRef] [PubMed]
  63. Salah, A.; Li, Y.; Wang, H.; Qi, N.; Wu, Y. Macrophages as a Double-Edged Weapon: The Use of Macrophages in Cancer Immunotherapy and Understanding the Cross-Talk Between Macrophages and Cancer. DNA Cell Biol. 2021, 40, 429–440. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, J.; Denu, R.A.; Dollar, B.A.; Escalante, L.E.; Kuether, J.P.; Callander, N.S.; Asimakopoulos, F.; Hematti, P. Macrophages and mesenchymal stromal cells support survival and proliferation of multiple myeloma cells. Br. J. Haematol. 2012, 158, 336–346. [Google Scholar] [CrossRef] [PubMed]
  65. Ramachandran, I.R.; Martner, A.; Pisklakova, A.; Condamine, T.; Chase, T.; Vogl, T.; Roth, J.; Gabrilovich, D.; Nefedova, Y. Myeloid-derived suppressor cells regulate growth of multiple myeloma by inhibiting T cells in bone marrow. J. Immunol. 2013, 190, 3815–3823. [Google Scholar] [CrossRef] [PubMed]
  66. Favaloro, J.; Liyadipitiya, T.; Brown, R.; Yang, S.; Suen, H.; Woodland, N.; Nassif, N.; Hart, D.; Fromm, P.; Weatherburn, C.; et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk. Lymphoma 2014, 55, 2893–2900. [Google Scholar] [CrossRef] [PubMed]
  67. Botta, C.; Gulla, A.; Correale, P.; Tagliaferri, P.; Tassone, P. Myeloid-derived suppressor cells in multiple myeloma: Pre-clinical research and translational opportunities. Front. Oncol. 2014, 4, 348. [Google Scholar] [CrossRef]
  68. Schmielau, J.; Finn, O.J. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients. Cancer Res. 2001, 61, 4756–4760. [Google Scholar] [PubMed]
  69. Mantovani, G.; Maccio, A.; Madeddu, C.; Mura, L.; Gramignano, G.; Lusso, M.R.; Massa, E.; Mocci, M.; Serpe, R. Antioxidant agents are effective in inducing lymphocyte progression through cell cycle in advanced cancer patients: Assessment of the most important laboratory indexes of cachexia and oxidative stress. J. Mol. Med. 2003, 81, 664–673. [Google Scholar] [CrossRef]
  70. Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar] [CrossRef]
  71. Serafini, P.; Mgebroff, S.; Noonan, K.; Borrello, I. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res. 2008, 68, 5439–5449. [Google Scholar] [CrossRef]
  72. Pan, P.Y.; Ma, G.; Weber, K.J.; Ozao-Choy, J.; Wang, G.; Yin, B.; Divino, C.M.; Chen, S.H. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010, 70, 99–108. [Google Scholar] [CrossRef] [PubMed]
  73. Abramson, H.N. B-Cell Maturation Antigen (BCMA) as a Target for New Drug Development in Relapsed and/or Refractory Multiple Myeloma. Int. J. Mol. Sci. 2020, 21, 5192. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, Z.; Zhang, L.; Wang, H.; Xiong, S.; Li, Y.; Tao, Q.; Xiao, W.; Qin, H.; Wang, Y.; Zhai, Z. Tumor-induced CD14+HLA-DR−/low myeloid-derived suppressor cells correlate with tumor progression and outcome of therapy in multiple myeloma patients. Cancer Immunol. Immunother. 2015, 64, 389–399. [Google Scholar] [CrossRef] [PubMed]
  75. Gorgun, G.T.; Whitehill, G.; Anderson, J.L.; Hideshima, T.; Maguire, C.; Laubach, J.; Raje, N.; Munshi, N.C.; Richardson, P.G.; Anderson, K.C. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood 2013, 121, 2975–2987. [Google Scholar] [CrossRef] [PubMed]
  76. Terabe, M.; Khanna, C.; Bose, S.; Melchionda, F.; Mendoza, A.; Mackall, C.L.; Helman, L.J.; Berzofsky, J.A. CD1d-restricted natural killer T cells can down-regulate tumor immunosurveillance independent of interleukin-4 receptor-signal transducer and activator of transcription 6 or transforming growth factor-beta. Cancer Res. 2006, 66, 3869–3875. [Google Scholar] [CrossRef] [PubMed]
  77. Harrison, S.J.; Cook, G. Immunotherapy in multiple myeloma—Possibility or probability? Br. J. Haematol. 2005, 130, 344–362. [Google Scholar] [CrossRef] [PubMed]
  78. Kronenberg, M. Toward an understanding of NKT cell biology: Progress and paradoxes. Annu. Rev. Immunol. 2005, 23, 877–900. [Google Scholar] [CrossRef] [PubMed]
  79. MacDonald, H.R. NK1.1+ T cell receptor-alpha/beta+ cells: New clues to their origin, specificity, and function. J. Exp. Med. 1995, 182, 633–638. [Google Scholar] [CrossRef]
  80. Dhodapkar, M.V.; Richter, J. Harnessing natural killer T (NKT) cells in human myeloma: Progress and challenges. Clin. Immunol. 2011, 140, 160–166. [Google Scholar] [CrossRef]
  81. Dhodapkar, M.V.; Geller, M.D.; Chang, D.H.; Shimizu, K.; Fujii, S.; Dhodapkar, K.M.; Krasovsky, J. A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J. Exp. Med. 2003, 197, 1667–1676. [Google Scholar] [CrossRef]
  82. Spanoudakis, E.; Hu, M.; Naresh, K.; Terpos, E.; Melo, V.; Reid, A.; Kotsianidis, I.; Abdalla, S.; Rahemtulla, A.; Karadimitris, A. Regulation of multiple myeloma survival and progression by CD1d. Blood 2009, 113, 2498–2507. [Google Scholar] [CrossRef] [PubMed]
  83. Jurczyszyn, A.; Gdula-Argasinska, J.; Kosmaczewska, A.; Skotnicki, A.B. The role of the bone marrow microenvironment in the pathogenesis of multiple myeloma. Postepy Hig. Med. Dosw. 2015, 69, 521–533. [Google Scholar] [CrossRef]
  84. Avella Patino, D.M.; Radhakrishnan, V.; Suvilesh, K.N.; Manjunath, Y.; Li, G.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Warren, W.C.; Kaifi, J.T.; Mitchem, J.B. Epigenetic Regulation of Cancer Immune Cells. Semin. Cancer Biol. 2022, 83, 377–383. [Google Scholar] [CrossRef] [PubMed]
  85. Bodey, B.; Siegel, S.E.; Kaiser, H.E. Antigen presentation by dendritic cells and their significance in antineoplastic immunotherapy. In Vivo 2004, 18, 81–100. [Google Scholar] [PubMed]
  86. Garcia De Vinuesa, C.; Gulbranson-Judge, A.; Khan, M.; O’Leary, P.; Cascalho, M.; Wabl, M.; Klaus, G.G.; Owen, M.J.; MacLennan, I.C. Dendritic cells associated with plasmablast survival. Eur. J. Immunol. 1999, 29, 3712–3721. [Google Scholar] [CrossRef]
  87. Jego, G.; Palucka, A.K.; Blanck, J.P.; Chalouni, C.; Pascual, V.; Banchereau, J. Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity 2003, 19, 225–234. [Google Scholar] [CrossRef] [PubMed]
  88. Brimnes, M.K.; Svane, I.M.; Johnsen, H.E. Impaired functionality and phenotypic profile of dendritic cells from patients with multiple myeloma. Clin. Exp. Immunol. 2006, 144, 76–84. [Google Scholar] [CrossRef]
  89. Leone, P.; Berardi, S.; Frassanito, M.A.; Ria, R.; De Re, V.; Cicco, S.; Battaglia, S.; Ditonno, P.; Dammacco, F.; Vacca, A.; et al. Dendritic cells accumulate in the bone marrow of myeloma patients where they protect tumor plasma cells from CD8+ T-cell killing. Blood 2015, 126, 1443–1451. [Google Scholar] [CrossRef]
  90. Schettini, J.; Mukherjee, P. Physiological role of plasmacytoid dendritic cells and their potential use in cancer immunity. Clin. Dev. Immunol. 2008, 2008, 106321. [Google Scholar] [CrossRef]
  91. Chauhan, D.; Singh, A.V.; Brahmandam, M.; Carrasco, R.; Bandi, M.; Hideshima, T.; Bianchi, G.; Podar, K.; Tai, Y.T.; Mitsiades, C.; et al. Functional interaction of plasmacytoid dendritic cells with multiple myeloma cells: A therapeutic target. Cancer Cell 2009, 16, 309–323. [Google Scholar] [CrossRef]
  92. Bi, E.; Li, R.; Bover, L.C.; Li, H.; Su, P.; Ma, X.; Huang, C.; Wang, Q.; Liu, L.; Yang, M.; et al. E-cadherin expression on multiple myeloma cells activates tumor-promoting properties in plasmacytoid DCs. J. Clin. Investig. 2018, 128, 4821–4831. [Google Scholar] [CrossRef]
  93. Pessoa de Magalhaes, R.J.; Vidriales, M.B.; Paiva, B.; Fernandez-Gimenez, C.; Garcia-Sanz, R.; Mateos, M.V.; Gutierrez, N.C.; Lecrevisse, Q.; Blanco, J.F.; Hernandez, J.; et al. Analysis of the immune system of multiple myeloma patients achieving long-term disease control by multidimensional flow cytometry. Haematologica 2013, 98, 79–86. [Google Scholar] [CrossRef] [PubMed]
  94. Brimnes, M.K.; Vangsted, A.J.; Knudsen, L.M.; Gimsing, P.; Gang, A.O.; Johnsen, H.E.; Svane, I.M. Increased level of both CD4+FOXP3+ regulatory T cells and CD14+HLA-DR/low myeloid-derived suppressor cells and decreased level of dendritic cells in patients with multiple myeloma. Scand. J. Immunol. 2010, 72, 540–547. [Google Scholar] [CrossRef]
  95. Bascones-Martinez, A.; Mattila, R.; Gomez-Font, R.; Meurman, J.H. Immunomodulatory drugs: Oral and systemic adverse effects. Med. Oral. Patol. Oral. Cir. Bucal 2014, 19, e24–e31. [Google Scholar] [CrossRef]
  96. Holstein, S.A.; McCarthy, P.L. Immunomodulatory Drugs in Multiple Myeloma: Mechanisms of Action and Clinical Experience. Drugs 2017, 77, 505–520. [Google Scholar] [CrossRef]
  97. Quach, H.; Ritchie, D.; Stewart, A.K.; Neeson, P.; Harrison, S.; Smyth, M.J.; Prince, H.M. Mechanism of action of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia 2010, 24, 22–32. [Google Scholar] [CrossRef]
  98. Saltarella, I.; Altamura, C.; Campanale, C.; Laghetti, P.; Vacca, A.; Frassanito, M.A.; Desaphy, J.F. Anti-Angiogenic Activity of Drugs in Multiple Myeloma. Cancers 2023, 15, 1990. [Google Scholar] [CrossRef]
  99. Asatsuma-Okumura, T.; Ito, T.; Handa, H. Molecular Mechanisms of the Teratogenic Effects of Thalidomide. Pharmaceuticals 2020, 13, 95. [Google Scholar] [CrossRef]
  100. Singhal, S.; Mehta, J.; Desikan, R.; Ayers, D.; Roberson, P.; Eddlemon, P.; Munshi, N.; Anaissie, E.; Wilson, C.; Dhodapkar, M.; et al. Antitumor activity of thalidomide in refractory multiple myeloma. N. Engl. J. Med. 1999, 341, 1565–1571. [Google Scholar] [CrossRef]
  101. Palumbo, A.; Facon, T.; Sonneveld, P.; Blade, J.; Offidani, M.; Gay, F.; Moreau, P.; Waage, A.; Spencer, A.; Ludwig, H.; et al. Thalidomide for treatment of multiple myeloma: 10 years later. Blood 2008, 111, 3968–3977. [Google Scholar] [CrossRef]
  102. Escoubet-Lozach, L.; Lin, I.L.; Jensen-Pergakes, K.; Brady, H.A.; Gandhi, A.K.; Schafer, P.H.; Muller, G.W.; Worland, P.J.; Chan, K.W.; Verhelle, D. Pomalidomide and lenalidomide induce p21 WAF-1 expression in both lymphoma and multiple myeloma through a LSD1-mediated epigenetic mechanism. Cancer Res. 2009, 69, 7347–7356. [Google Scholar] [CrossRef] [PubMed]
  103. Hideshima, T.; Chauhan, D.; Shima, Y.; Raje, N.; Davies, F.E.; Tai, Y.T.; Treon, S.P.; Lin, B.; Schlossman, R.L.; Richardson, P.; et al. Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy. Blood 2000, 96, 2943–2950. [Google Scholar] [CrossRef] [PubMed]
  104. Niesvizky, R.; Jayabalan, D.S.; Christos, P.J.; Furst, J.R.; Naib, T.; Ely, S.; Jalbrzikowski, J.; Pearse, R.N.; Zafar, F.; Pekle, K.; et al. BiRD (Biaxin [clarithromycin]/Revlimid [lenalidomide]/dexamethasone) combination therapy results in high complete- and overall-response rates in treatment-naive symptomatic multiple myeloma. Blood 2008, 111, 1101–1109. [Google Scholar] [CrossRef] [PubMed]
  105. McCarthy, P.L.; Owzar, K.; Hofmeister, C.C.; Hurd, D.D.; Hassoun, H.; Richardson, P.G.; Giralt, S.; Stadtmauer, E.A.; Weisdorf, D.J.; Vij, R.; et al. Lenalidomide after stem-cell transplantation for multiple myeloma. N. Engl. J. Med. 2012, 366, 1770–1781. [Google Scholar] [CrossRef] [PubMed]
  106. Lacy, M.Q.; Tefferi, A. Pomalidomide therapy for multiple myeloma and myelofibrosis: An update. Leuk. Lymphoma 2011, 52, 560–566. [Google Scholar] [CrossRef] [PubMed]
  107. Frampton, J.E. Isatuximab: A Review of Its Use in Multiple Myeloma. Target. Oncol. 2021, 16, 675–686. [Google Scholar] [CrossRef] [PubMed]
  108. Yamanaka, S.; Murai, H.; Saito, D.; Abe, G.; Tokunaga, E.; Iwasaki, T.; Takahashi, H.; Takeda, H.; Suzuki, T.; Shibata, N.; et al. Thalidomide and its metabolite 5-hydroxythalidomide induce teratogenicity via the cereblon neosubstrate PLZF. EMBO J. 2021, 40, e105375. [Google Scholar] [CrossRef] [PubMed]
  109. Merz, M.; Dechow, T.; Scheytt, M.; Schmidt, C.; Hackanson, B.; Knop, S. The clinical management of lenalidomide-based therapy in patients with newly diagnosed multiple myeloma. Ann. Hematol. 2020, 99, 1709–1725. [Google Scholar] [CrossRef]
  110. Dimopoulos, M.; Weisel, K.; Moreau, P.; Anderson, L.D., Jr.; White, D.; San-Miguel, J.; Sonneveld, P.; Engelhardt, M.; Jenner, M.; Corso, A.; et al. Pomalidomide, bortezomib, and dexamethasone for multiple myeloma previously treated with lenalidomide (OPTIMISMM): Outcomes by prior treatment at first relapse. Leukemia 2021, 35, 1722–1731. [Google Scholar] [CrossRef]
  111. Richardson, P.G.; Trudel, S.; Popat, R.; Mateos, M.V.; Vangsted, A.J.; Ramasamy, K.; Martinez-Lopez, J.; Quach, H.; Orlowski, R.Z.; Arnao, M.; et al. Mezigdomide plus Dexamethasone in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2023, 389, 1009–1022. [Google Scholar] [CrossRef]
  112. Field-Smith, A.; Morgan, G.J.; Davies, F.E. Bortezomib (Velcadetrade mark) in the Treatment of Multiple Myeloma. Ther. Clin. Risk Manag. 2006, 2, 271–279. [Google Scholar] [CrossRef] [PubMed]
  113. Kouroukis, T.C.; Baldassarre, F.G.; Haynes, A.E.; Imrie, K.; Reece, D.E.; Cheung, M.C. Bortezomib in multiple myeloma: Systematic review and clinical considerations. Curr. Oncol. 2014, 21, e573–e603. [Google Scholar] [CrossRef] [PubMed]
  114. Jayaweera, S.P.E.; Wanigasinghe Kanakanamge, S.P.; Rajalingam, D.; Silva, G.N. Carfilzomib: A Promising Proteasome Inhibitor for the Treatment of Relapsed and Refractory Multiple Myeloma. Front. Oncol. 2021, 11, 740796. [Google Scholar] [CrossRef] [PubMed]
  115. Muz, B.; Ghazarian, R.N.; Ou, M.; Luderer, M.J.; Kusdono, H.D.; Azab, A.K. Spotlight on ixazomib: Potential in the treatment of multiple myeloma. Drug Des. Dev. Ther. 2016, 10, 217–226. [Google Scholar] [CrossRef]
  116. Rajkumar, S.V. Multiple myeloma: 2022 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2022, 97, 1086–1107. [Google Scholar] [CrossRef] [PubMed]
  117. McKenzie, F.; Gazze, G.; Hewitt, J.; Kolm, K.; Pollock, D.; Rowland, S.; Crosbie, T. Canadian perspectives in multiple myeloma on the use of steroids in clinical practice based on patient and healthcare provider interviews. Front. Oncol. 2022, 12, 1061417. [Google Scholar] [CrossRef] [PubMed]
  118. Mateos, M.V.; Dimopoulos, M.A.; Cavo, M.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; Kaplan, P.; et al. Daratumumab plus Bortezomib, Melphalan, and Prednisone for Untreated Myeloma. N. Engl. J. Med. 2018, 378, 518–528. [Google Scholar] [CrossRef] [PubMed]
  119. Costa, B.A.; Mouhieddine, T.H.; Ortiz, R.J.; Richter, J. Revisiting the role of alkylating agents in multiple myeloma: Up-to-date evidence and future perspectives. Crit. Rev. Oncol. Hematol. 2023, 187, 104040. [Google Scholar] [CrossRef]
  120. Lu, X.; Ding, Z.C.; Cao, Y.; Liu, C.; Habtetsion, T.; Yu, M.; Lemos, H.; Salman, H.; Xu, H.; Mellor, A.L.; et al. Alkylating agent melphalan augments the efficacy of adoptive immunotherapy using tumor-specific CD4+ T cells. J. Immunol. 2015, 194, 2011–2021. [Google Scholar] [CrossRef]
  121. Kaiser, M.F.; Hall, A.; Walker, K.; Sherborne, A.; De Tute, R.M.; Newnham, N.; Roberts, S.; Ingleson, E.; Bowles, K.; Garg, M.; et al. Daratumumab, Cyclophosphamide, Bortezomib, Lenalidomide, and Dexamethasone as Induction and Extended Consolidation Improves Outcome in Ultra-High-Risk Multiple Myeloma. J. Clin. Oncol. 2023, 41, 3945–3955. [Google Scholar] [CrossRef]
  122. Sidana, S.; Hosoya, H.; Jensen, A.; Liu, L.; Goyal, A.; Hovanky, V.; Sahaf, B.; Bharadwaj, S.; Latchford, T.; Arai, S.; et al. Bendamustine vs. fludarabine/cyclophosphamide lymphodepletion prior to BCMA CAR-T cell therapy in multiple myeloma. Blood Cancer J. 2023, 13, 158. [Google Scholar] [CrossRef] [PubMed]
  123. Gentile, M.; Vigna, E.; Recchia, A.G.; Morabito, L.; Mendicino, F.; Giagnuolo, G.; Morabito, F. Bendamustine in multiple myeloma. Eur. J. Haematol. 2015, 95, 377–388. [Google Scholar] [CrossRef] [PubMed]
  124. Afifi, S.; Michael, A.; Lesokhin, A. Immunotherapy: A New Approach to Treating Multiple Myeloma with Daratumumab and Elotuzumab. Ann. Pharmacother. 2016, 50, 555–568. [Google Scholar] [CrossRef]
  125. Jadoon, Y.; Siddiqui, M.A. Immunotherapy in multiple myeloma. Cancer Treat. Res. Commun. 2021, 29, 100468. [Google Scholar] [CrossRef] [PubMed]
  126. Nooka, A.K.; Kaufman, J.L.; Hofmeister, C.C.; Joseph, N.S.; Heffner, T.L.; Gupta, V.A.; Sullivan, H.C.; Neish, A.S.; Dhodapkar, M.V.; Lonial, S. Daratumumab in multiple myeloma. Cancer 2019, 125, 2364–2382. [Google Scholar] [CrossRef] [PubMed]
  127. Phipps, C.; Chen, Y.; Gopalakrishnan, S.; Tan, D. Daratumumab and its potential in the treatment of multiple myeloma: Overview of the preclinical and clinical development. Ther. Adv. Hematol. 2015, 6, 120–127. [Google Scholar] [CrossRef] [PubMed]
  128. Dimopoulos, K.; Gimsing, P.; Gronbaek, K. The role of epigenetics in the biology of multiple myeloma. Blood Cancer J. 2014, 4, e207. [Google Scholar] [CrossRef]
  129. Pan, D.; Mouhieddine, T.H.; Upadhyay, R.; Casasanta, N.; Lee, A.; Zubizarreta, N.; Moshier, E.; Richter, J. Outcomes with panobinostat in heavily pretreated multiple myeloma patients. Semin. Oncol. 2023, 50, 40–48. [Google Scholar] [CrossRef] [PubMed]
  130. Corral, L.G.; Haslett, P.A.; Muller, G.W.; Chen, R.; Wong, L.M.; Ocampo, C.J.; Patterson, R.T.; Stirling, D.I.; Kaplan, G. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. J. Immunol. 1999, 163, 380–386. [Google Scholar] [CrossRef] [PubMed]
  131. Gupta, D.; Treon, S.P.; Shima, Y.; Hideshima, T.; Podar, K.; Tai, Y.T.; Lin, B.; Lentzsch, S.; Davies, F.E.; Chauhan, D.; et al. Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: Therapeutic applications. Leukemia 2001, 15, 1950–1961. [Google Scholar] [CrossRef]
  132. Zhu, Y.X.; Kortuem, K.M.; Stewart, A.K. Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk. Lymphoma 2013, 54, 683–687. [Google Scholar] [CrossRef] [PubMed]
  133. Breitkreutz, I.; Raab, M.S.; Vallet, S.; Hideshima, T.; Raje, N.; Mitsiades, C.; Chauhan, D.; Okawa, Y.; Munshi, N.C.; Richardson, P.G.; et al. Lenalidomide inhibits osteoclastogenesis, survival factors and bone-remodeling markers in multiple myeloma. Leukemia 2008, 22, 1925–1932. [Google Scholar] [CrossRef]
  134. Giannopoulos, K.; Kaminska, W.; Hus, I.; Dmoszynska, A. The frequency of T regulatory cells modulates the survival of multiple myeloma patients: Detailed characterisation of immune status in multiple myeloma. Br. J. Cancer 2012, 106, 546–552. [Google Scholar] [CrossRef] [PubMed]
  135. Galustian, C.; Meyer, B.; Labarthe, M.C.; Dredge, K.; Klaschka, D.; Henry, J.; Todryk, S.; Chen, R.; Muller, G.; Stirling, D.; et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol. Immunother. 2009, 58, 1033–1045. [Google Scholar] [CrossRef] [PubMed]
  136. Zhu, D.; Corral, L.G.; Fleming, Y.W.; Stein, B. Immunomodulatory drugs Revlimid (lenalidomide) and CC-4047 induce apoptosis of both hematological and solid tumor cells through NK cell activation. Cancer Immunol. Immunother. 2008, 57, 1849–1859. [Google Scholar] [CrossRef] [PubMed]
  137. Wu, L.; Adams, M.; Carter, T.; Chen, R.; Muller, G.; Stirling, D.; Schafer, P.; Bartlett, J.B. lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells. Clin. Cancer Res. 2008, 14, 4650–4657. [Google Scholar] [CrossRef] [PubMed]
  138. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
  139. D’Agostino, M.; Boccadoro, M.; Smith, E.L. Novel Immunotherapies for Multiple Myeloma. Curr. Hematol. Malig. Rep. 2017, 12, 344–357. [Google Scholar] [CrossRef] [PubMed]
  140. Lao, Y.; Shen, D.; Zhang, W.; He, R.; Jiang, M. Immune Checkpoint Inhibitors in Cancer Therapy—How to Overcome Drug Resistance? Cancers 2022, 14, 3575. [Google Scholar] [CrossRef]
  141. Lee, M.Y.; Park, C.J.; Cho, Y.U.; You, E.; Jang, S.; Seo, E.J.; Lee, J.H.; Yoon, D.H.; Suh, C. Immune Checkpoint Programmed Cell Death Protein-1 (PD-1) Expression on Bone Marrow T Cell Subsets in Patients With Plasma Cell Myeloma. Ann. Lab. Med. 2021, 41, 259–267. [Google Scholar] [CrossRef]
  142. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar] [PubMed]
  143. Chambers, C.A.; Kuhns, M.S.; Allison, J.P. Cytotoxic T lymphocyte antigen-4 (CTLA-4) regulates primary and secondary peptide-specific CD4+ T cell responses. Proc. Natl. Acad. Sci. USA 1999, 96, 8603–8608. [Google Scholar] [CrossRef] [PubMed]
  144. Braga, W.M.; da Silva, B.R.; de Carvalho, A.C.; Maekawa, Y.H.; Bortoluzzo, A.B.; Rizzatti, E.G.; Atanackovic, D.; Colleoni, G.W. FOXP3 and CTLA4 overexpression in multiple myeloma bone marrow as a sign of accumulation of CD4+ T regulatory cells. Cancer Immunol. Immunother. 2014, 63, 1189–1197. [Google Scholar] [CrossRef] [PubMed]
  145. Zanwar, S.; Nandakumar, B.; Kumar, S. Immune-based therapies in the management of multiple myeloma. Blood Cancer J. 2020, 10, 84. [Google Scholar] [CrossRef] [PubMed]
  146. Mateos, M.V.; Blacklock, H.; Schjesvold, F.; Oriol, A.; Simpson, D.; George, A.; Goldschmidt, H.; Larocca, A.; Chanan-Khan, A.; Sherbenou, D.; et al. Pembrolizumab plus pomalidomide and dexamethasone for patients with relapsed or refractory multiple myeloma (KEYNOTE-183): A randomised, open-label, phase 3 trial. Lancet Haematol. 2019, 6, e459–e469. [Google Scholar] [CrossRef] [PubMed]
  147. Atrash, S.; Ali, S.A.; Usmani, S.Z. Chimeric Antigen Receptor T-cell Therapy for Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2021, 21, 21–34. [Google Scholar] [CrossRef]
  148. Choi, T.; Kang, Y. Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma. Pharmacol. Ther. 2022, 232, 108007. [Google Scholar] [CrossRef]
  149. Brentjens, R.J.; Latouche, J.B.; Santos, E.; Marti, F.; Gong, M.C.; Lyddane, C.; King, P.D.; Larson, S.; Weiss, M.; Riviere, I.; et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med. 2003, 9, 279–286. [Google Scholar] [CrossRef]
  150. Kochenderfer, J.N.; Feldman, S.A.; Zhao, Y.; Xu, H.; Black, M.A.; Morgan, R.A.; Wilson, W.H.; Rosenberg, S.A. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J. Immunother. 2009, 32, 689–702. [Google Scholar] [CrossRef]
  151. Jena, B.; Moyes, J.S.; Huls, H.; Cooper, L.J. Driving CAR-based T-cell therapy to success. Curr. Hematol. Malig. Rep. 2014, 9, 50–56. [Google Scholar] [CrossRef]
  152. O’Connor, B.P.; Raman, V.S.; Erickson, L.D.; Cook, W.J.; Weaver, L.K.; Ahonen, C.; Lin, L.L.; Mantchev, G.T.; Bram, R.J.; Noelle, R.J. BCMA is essential for the survival of long-lived bone marrow plasma cells. J. Exp. Med. 2004, 199, 91–98. [Google Scholar] [CrossRef] [PubMed]
  153. Sanchez, E.; Li, M.; Kitto, A.; Li, J.; Wang, C.S.; Kirk, D.T.; Yellin, O.; Nichols, C.M.; Dreyer, M.P.; Ahles, C.P.; et al. Serum B-cell maturation antigen is elevated in multiple myeloma and correlates with disease status and survival. Br. J. Haematol. 2012, 158, 727–738. [Google Scholar] [CrossRef]
  154. Moreaux, J.; Legouffe, E.; Jourdan, E.; Quittet, P.; Reme, T.; Lugagne, C.; Moine, P.; Rossi, J.F.; Klein, B.; Tarte, K. BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone. Blood 2004, 103, 3148–3157. [Google Scholar] [CrossRef] [PubMed]
  155. Ali, S.A.; Shi, V.; Maric, I.; Wang, M.; Stroncek, D.F.; Rose, J.J.; Brudno, J.N.; Stetler-Stevenson, M.; Feldman, S.A.; Hansen, B.G.; et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 2016, 128, 1688–1700. [Google Scholar] [CrossRef] [PubMed]
  156. Berdeja, J.G.; Madduri, D.; Usmani, S.Z.; Jakubowiak, A.; Agha, M.; Cohen, A.D.; Stewart, A.K.; Hari, P.; Htut, M.; Lesokhin, A.; et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study. Lancet 2021, 398, 314–324. [Google Scholar] [CrossRef] [PubMed]
  157. Clinicaltrials.gov. A Study of Whether Ide-cel (bb2121) Can Be Made from People with Multiple Myeloma Who Have Had a Hematopoietic Cell Transplant [NCT05393804]. Available online: https://www.clinicaltrials.gov/study/NCT05393804?term=NCT05393804&rank=1 (accessed on 25 February 2024).
  158. Raje, N.; Berdeja, J.; Lin, Y.; Siegel, D.; Jagannath, S.; Madduri, D.; Liedtke, M.; Rosenblatt, J.; Maus, M.V.; Turka, A.; et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 380, 1726–1737. [Google Scholar] [CrossRef] [PubMed]
  159. Munshi, N.C.; Anderson, L.D., Jr.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716. [Google Scholar] [CrossRef] [PubMed]
  160. San-Miguel, J.; Dhakal, B.; Yong, K.; Spencer, A.; Anguille, S.; Mateos, M.V.; Fernandez de Larrea, C.; Martinez-Lopez, J.; Moreau, P.; Touzeau, C.; et al. Cilta-cel or Standard Care in Lenalidomide-Refractory Multiple Myeloma. N. Engl. J. Med. 2023, 389, 335–347. [Google Scholar] [CrossRef] [PubMed]
  161. Dhakal, B.; Yong, K.; Harrison, S.J.; Mateos, M.-V.; Moreau, P.; van de Donk, N.W.C.J.; Sidana, S.; Popat, R.; Lendvai, N.; Lonardi, C.; et al. First phase 3 results from CARTITUDE-4: Cilta-cel versus standard of care (PVd or DPd) in lenalidomide-refractory multiple myeloma. J. Clin. Oncol. 2023, 41, LBA106. [Google Scholar] [CrossRef]
  162. Zhao, W.H.; Liu, J.; Wang, B.Y.; Chen, Y.X.; Cao, X.M.; Yang, Y.; Zhang, Y.L.; Wang, F.X.; Zhang, P.Y.; Lei, B.; et al. A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J. Hematol. Oncol. 2018, 11, 141. [Google Scholar] [CrossRef]
  163. Mailankody, S.; Htut, M.; Lee, K.P.; Bensinger, W.; Devries, T.; Piasecki, J.; Ziyad, S.; Blake, M.; Byon, J.; Jakubowiak, A. JCARH125, Anti-BCMA CAR T-cell Therapy for Relapsed/Refractory Multiple Myeloma: Initial Proof of Concept Results from a Phase 1/2 Multicenter Study (EVOLVE). Blood 2018, 132, 957. [Google Scholar] [CrossRef]
  164. Mailankody, S.; Jakubowiak, A.J.; Htut, M.; Costa, L.J.; Lee, K.P.; Ganguly, S.; Kaufman, J.L.; Siegel, D.S.D.; Bensinger, W.; Cota, M.; et al. Orvacabtagene autoleucel (orva-cel), a B-cell maturation antigen (BCMA)-directed CAR T cell therapy for patients (pts) with relapsed/refractory multiple myeloma (RRMM): Update of the phase 1/2 EVOLVE study (NCT03430011). J. Clin. Oncol. 2020, 38, 8504. [Google Scholar] [CrossRef]
  165. Frigault, M.J.; Bishop, M.R.; Rosenblatt, J.; O’Donnell, E.K.; Raje, N.; Cook, D.; Yee, A.J.; Logan, E.; Avigan, D.E.; Jakubowiak, A.; et al. Phase 1 study of CART-ddBCMA for the treatment of subjects with relapsed and refractory multiple myeloma. Blood Adv. 2023, 7, 768–777. [Google Scholar] [CrossRef] [PubMed]
  166. 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]
  167. Wang, X.; Pan, B.; Huang, H.; Xu, K. Non-BCMA targeted CAR-T cell therapies for multiple myeloma. ImmunoMedicine 2021, 1, e1030. [Google Scholar] [CrossRef]
  168. Yan, Z.; Cao, J.; Cheng, H.; Qiao, J.; Zhang, H.; Wang, Y.; Shi, M.; Lan, J.; Fei, X.; Jin, L.; et al. A combination of humanised anti-CD19 and anti-BCMA CAR T cells in patients with relapsed or refractory multiple myeloma: A single-arm, phase 2 trial. Lancet Haematol. 2019, 6, e521–e529. [Google Scholar] [CrossRef] [PubMed]
  169. Kodama, T.; Kochi, Y.; Nakai, W.; Mizuno, H.; Baba, T.; Habu, K.; Sawada, N.; Tsunoda, H.; Shima, T.; Miyawaki, K.; et al. Anti-GPRC5D/CD3 Bispecific T-Cell-Redirecting Antibody for the Treatment of Multiple Myeloma. Mol. Cancer Ther. 2019, 18, 1555–1564. [Google Scholar] [CrossRef]
  170. Sun, C.; Mahendravada, A.; Ballard, B.; Kale, B.; Ramos, C.; West, J.; Maguire, T.; McKay, K.; Lichtman, E.; Tuchman, S.; et al. Safety and efficacy of targeting CD138 with a chimeric antigen receptor for the treatment of multiple myeloma. Oncotarget 2019, 10, 2369–2383. [Google Scholar] [CrossRef]
  171. Thomas, R.; Al-Khadairi, G.; Roelands, J.; Hendrickx, W.; Dermime, S.; Bedognetti, D.; Decock, J. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front. Immunol. 2018, 9, 947. [Google Scholar] [CrossRef]
  172. Rapoport, A.P.; Stadtmauer, E.A.; Binder-Scholl, G.K.; Goloubeva, O.; Vogl, D.T.; Lacey, S.F.; Badros, A.Z.; Garfall, A.; Weiss, B.; Finklestein, J.; et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 2015, 21, 914–921. [Google Scholar] [CrossRef]
  173. Cannons, J.L.; Tangye, S.G.; Schwartzberg, P.L. SLAM family receptors and SAP adaptors in immunity. Annu. Rev. Immunol. 2011, 29, 665–705. [Google Scholar] [CrossRef]
  174. Dimopoulos, M.A.; Dytfeld, D.; Grosicki, S.; Moreau, P.; Takezako, N.; Hori, M.; Leleu, X.; LeBlanc, R.; Suzuki, K.; Raab, M.S.; et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2018, 379, 1811–1822. [Google Scholar] [CrossRef]
  175. Curio, S.; Jonsson, G.; Marinovic, S. A summary of current NKG2D-based CAR clinical trials. Immunother. Adv. 2021, 1, ltab018. [Google Scholar] [CrossRef]
  176. Spear, P.; Wu, M.R.; Sentman, M.L.; Sentman, C.L. NKG2D ligands as therapeutic targets. Cancer Immun. 2013, 13, 8. [Google Scholar]
  177. Lanier, L.L. NKG2D Receptor and Its Ligands in Host Defense. Cancer Immunol. Res. 2015, 3, 575–582. [Google Scholar] [CrossRef]
  178. Spear, P.; Barber, A.; Rynda-Apple, A.; Sentman, C.L. NKG2D CAR T-cell therapy inhibits the growth of NKG2D ligand heterogeneous tumors. Immunol. Cell Biol. 2013, 91, 435–440. [Google Scholar] [CrossRef]
  179. Garfall, A.L.; Stadtmauer, E.A.; Hwang, W.T.; Lacey, S.F.; Melenhorst, J.J.; Krevvata, M.; Carroll, M.P.; Matsui, W.H.; Wang, Q.; Dhodapkar, M.V.; et al. Anti-CD19 CAR T cells with high-dose melphalan and autologous stem cell transplantation for refractory multiple myeloma. JCI Insight 2018, 3, e120505. [Google Scholar] [CrossRef]
  180. van der Schans, J.J.; van de Donk, N.; Mutis, T. Dual Targeting to Overcome Current Challenges in Multiple Myeloma CAR T-Cell Treatment. Front. Oncol. 2020, 10, 1362. [Google Scholar] [CrossRef] [PubMed]
  181. Baumeister, S.H.; Murad, J.; Werner, L.; Daley, H.; Trebeden-Negre, H.; Gicobi, J.K.; Schmucker, A.; Reder, J.; Sentman, C.L.; Gilham, D.E.; et al. Phase I Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol. Res. 2019, 7, 100–112. [Google Scholar] [CrossRef] [PubMed]
  182. Sallman, D.A.; Brayer, J.B.; Poire, X.; Havelange, V.; Awada, A.; Lewalle, P.; Odunsi, K.; Wang, E.S.; Lonez, C.; Lequertier, T.; et al. Results from the Completed Dose-Escalation of the Hematological Arm of the Phase I Think Study Evaluating Multiple Infusions of NKG2D-Based CAR T-Cells As Standalone Therapy in Relapse/Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. Blood 2019, 134, 3826. [Google Scholar] [CrossRef]
  183. Bal, S.; Kocoglu, M.H.; Nadeem, O.; Htut, M.; Gregory, T.; Anderson, L.D.; Costa, L.J.; Buchholz, T.J.; Ziyad, S.; Li, M.; et al. Clinical Activity of BMS-986393 (CC-95266), a G Protein-Coupled Receptor Class C Group 5 Member D (GPRC5D)-Targeted Chimeric Antigen Receptor (CAR) T Cell Therapy, in Patients with Relapsed and/or Refractory (R/R) Multiple Myeloma (MM): First Results from a Phase 1, Multicenter, Open-Label Study. Blood 2022, 140, 883–885. [Google Scholar] [CrossRef]
  184. Lin, Q.; Zhao, J.; Song, Y.; Liu, D. Recent updates on CAR T clinical trials for multiple myeloma. Mol. Cancer 2019, 18, 154. [Google Scholar] [CrossRef] [PubMed]
  185. Duan, D.; Wang, K.; Wei, C.; Feng, D.; Liu, Y.; He, Q.; Xu, X.; Wang, C.; Zhao, S.; Lv, L.; et al. The BCMA-Targeted Fourth-Generation CAR-T Cells Secreting IL-7 and CCL19 for Therapy of Refractory/Recurrent Multiple Myeloma. Front. Immunol. 2021, 12, 609421. [Google Scholar] [CrossRef] [PubMed]
  186. Clinicaltrials.gov. Safety Study of Anti LewisY Chimeric Antigen Receptor in Myeloma, Acute Myeloid Leukemia or Myelodysplastic Syndrome [NCT01716364]. Available online: https://www.clinicaltrials.gov/study/NCT01716364?term=NCT01716364&rank=1 (accessed on 25 February 2024).
  187. Clinicaltrials.gov. Study to Evaluate the Safety and Efficacy of Anti-CD38 CAR-T in Relapsed or Refractory Multiple Myeloma Patients [NCT03464916]. Available online: https://www.clinicaltrials.gov/study/NCT03464916?term=NCT03464916&rank=1 (accessed on 25 February 2024).
  188. Owusu, K.A.; Schiffer, M.; Perreault, S. Chimeric Antigen Receptor T Cells: Toxicity and Management Considerations. AACN Adv. Crit. Care 2022, 33, 301–307. [Google Scholar] [CrossRef]
  189. Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.A.; Wang, M.; Arnason, J.; Mehta, A.; Purev, E.; Maloney, D.G.; Andreadis, C.; et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet 2020, 396, 10254. [Google Scholar] [CrossRef]
  190. Xiao, X.; Huang, S.; Chen, S.; Wang, Y.; Sun, Q.; Xu, X.; Li, Y. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. J. Exp. Clin. Cancer Res. 2021, 40, 367. [Google Scholar] [CrossRef] [PubMed]
  191. Holtzman, N.G.; Xie, H.; Bentzen, S.; Kesari, V.; Bukhari, A.; El Chaer, F.; Lutfi, F.; Siglin, J.; Hutnick, E.; Gahres, N.; et al. Immune effector cell-associated neurotoxicity syndrome after chimeric antigen receptor T-cell therapy for lymphoma: Predictive biomarkers and clinical outcomes. Neuro Oncol. 2021, 23, 112–121. [Google Scholar] [CrossRef] [PubMed]
  192. Taefehshokr, S.; Parhizkar, A.; Hayati, S.; Mousapour, M.; Mahmoudpour, A.; Eleid, L.; Rahmanpour, D.; Fattahi, S.; Shabani, H.; Taefehshokr, N. Cancer immunotherapy: Challenges and limitations. Pathol. Res. Pract. 2022, 229, 153723. [Google Scholar] [CrossRef]
  193. Wu, J.; Cai, J. Dilemma and Challenge of Immunotherapy for Pancreatic Cancer. Dig. Dis. Sci. 2021, 66, 359–368. [Google Scholar] [CrossRef]
  194. Gupta, S.; Shukla, S. Limitations of Immunotherapy in Cancer. Cureus 2022, 14, e30856. [Google Scholar] [CrossRef]
  195. 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] [PubMed]
  196. Le Calvez, B.; Moreau, P.; Touzeau, C. Immune checkpoint inhibitors for the treatment of myeloma: Novel investigational options. Expert Opin. Investig. Drugs 2021, 30, 965–973. [Google Scholar] [CrossRef] [PubMed]
  197. Nadeem, O.; Tai, Y.T.; Anderson, K.C. Immunotherapeutic and Targeted Approaches in Multiple Myeloma. Immunotargets Ther. 2020, 9, 201–215. [Google Scholar] [CrossRef] [PubMed]
  198. Sheykhhasan, M.; Ahmadieh-Yazdi, A.; Vicidomini, R.; Poondla, N.; Tanzadehpanah, H.; Dirbaziyan, A.; Mahaki, H.; Manoochehri, H.; Kalhor, N.; Dama, P. CAR T therapies in multiple myeloma: Unleashing the future. Cancer Gene Ther. 2024. [Google Scholar] [CrossRef]
Figure 1. Graphical representation of different immune cells and their regulation in myeloma. Modulation of immune cell function by immunotherapy agents (IMiDs—immunomodulatory drugs; ICIs—immune checkpoint inhibitors) is used for multiple myeloma treatment.
Figure 1. Graphical representation of different immune cells and their regulation in myeloma. Modulation of immune cell function by immunotherapy agents (IMiDs—immunomodulatory drugs; ICIs—immune checkpoint inhibitors) is used for multiple myeloma treatment.
Life 14 00461 g001
Table 1. Drugs used in the treatment of multiple myeloma.
Table 1. Drugs used in the treatment of multiple myeloma.
DrugsMode of ActionRoute of
Administration
Overall Response Rate (ORR)References
Immunomodulatory Drugs
ThalidomideBinds to CRBN and triggers proteasomal degradation of IKZF1 and IKZF3, leading to direct anti-multiple myeloma activity and immunomodulatory and stromal cell effects.Oral28%[96,108]
LenalidomideBind CRBN, the substrate adaptor for the CRBN-CRL4 E3 ubiquitin ligase, and modulate the enzyme’s substrate specificity. These drugs induce the degradation of two lymphoid transcription factors, IKZF1 and IKZF3, leading to dramatic clinical efficacy in multiple myeloma.Oral25–27%[109]
PomalidomideDegrades the IKZF1 and IKZF3 proteins more potentially.Oral30–35%[110]
MezigdomideThe drug is designed to achieve rapid, potent, and deep degradation of Ikaros and Aiolos, key transcription factors in hematopoietic cell development and differentiation.Oral40%[111]
Proteasome inhibitors
BortezomibPrimarily acts through plasma cell depletion, reducing antibody production to T-dependent antigens.Intravenous (IV) or subcutaneous injection38%[112,113]
CarfilzomibThe drug irreversibly binds with the proteasome and inhibits its chymotrypsin-like activity.Intravenous infusion23%[114]
IxazomibReversibly binds and inhibits the chymotrypsin-like proteolytic (β5) site of the 20S proteasome, reducing the tumor progression. Oral78%[115]
Corticosteroids/Steroids
DexamethasoneAgonist to glucocorticoid receptor. It prevents white blood cells from migrating to locations where malignant myeloma cells are causing damage and reduces swelling and inflammation.Oral or intravenous90%[116]
PrednisoneAgonist to glucocorticoid receptor.Oral10%[117,118]
Alkylating drugs
MelphalanMelphalan induces inter- or infrastructural
DNA crosslinks and DNA–protein crosslinks. Increases
tumor-specific CD4 + T-cell responses and inhibits BM stromal cell expression of IL-6.
Intravenous (IV) and oral82%[119,120]
CyclophosphamideThe formation of DNA crosslinking alters cytokine expression in the BM microenvironment, causing MM cell death/apoptosis.Intravenous (IV) and oral58.1%[121]
BendamustineFormation of DNA crosslinking. Checkpoint controls inhibition, causing a mitotic catastrophe.Intravenous (IV)55%[122,123]
Monoclonal Antibodies
ElotuzumabDrug targets the signaling lymphocytic activation molecule F7 (SLAMF7). It directly activates natural killer cells, enhancing their ability to kill myeloma cells.Intravenous infusion79%[124,125]
DaratumumabIt works by targeting the protein CD38 on myeloma cells. It helps to slow or stop the progression of multiple myeloma.Intravenous infusion or subcutaneous injection81%[126,127]
IsatuximabAnti-CD38 monoclonal antibody exerts antimyeloma activity through several mechanisms of action, such as antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and direct induction of apoptotic cell death.Intravenous infusion23.8%[107]
Epigenetic inhibitors
PanobinostatIt is an HDAC inhibitor used in the treatment of relapsed and refractory multiple myeloma.Oral24.8%[128,129]
Table 2. Selected BCMA CAR-T clinical trials.
Table 2. Selected BCMA CAR-T clinical trials.
Trial NumberNamePhaseNumber of PatientsOverall Response Rate (ORR)SponsorsReferences
NCT03548207CARTITUDE-1211389%Janssen Research & Development, LLC[156]
NCT03338972 FCARH143112100%Fred Hutchinson Cancer Center[148]
NCT05393804bb21212 Recruiting NAMemorial Sloan Kettering Cancer Center[157]
NCT02658929 bb2121 or Ide-cel13385%Bluebird Bio and Celgene[158]
NCT02215967 CAR-BCMA12481%National Cancer Institute (NCI)[155]
NCT03361748 KarMMa 214073%Celgene[158,159]
NCT04181827 CARTITUDE-4 341976%Janssen Research & Development, LLC[160,161]
NCT03090659 LEGEND-2 1 5788%Nanjing Legend Biotech Co.[162]
NCT03430011 Orva-cel 1/24482%Juno Therapeutics, Celgene[163,164]
NCT04155749 NA 11360–75%Arcellx, Inc.[165]
Table 3. Selected non-BCMA-targeted CAR-T clinical trials.
Table 3. Selected non-BCMA-targeted CAR-T clinical trials.
Trial NumberNamePhaseNumber of PatientsSponsorsReferences
NCT02135406 CTL019112 University of Pennsylvania [179]
NCT03767725NA1 Recruiting Shenzhen Second People’s Hospital [73]
NCT03706547NA1UnknownPeng Liu[180]
NCT02203825NA16Celyad Oncology SA[181]
NCT03018405 THINK1 16 Celyad Oncology SA[182]
NCT04674813 CC-952661 21 Juno Therapeutics, a Subsidiary of Celgene [183]
NCT03710421 NA1 Recruiting City of Hope Medical Center [184]
NCT03778346 NA1 Recruiting The Sixth Affiliated Hospital of Wenzhou Medical University [185]
NCT01716364 NA1 6 Peter MacCallum Cancer Centre, Australia [186]
NCT03464916 CAR2 Anti-CD38 A21 Unknown Sorrento Therapeutics, Inc. [187]
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

Radhakrishnan, V.; Golla, U.; Kudva, A.K. Role of Immune Cells and Immunotherapy in Multiple Myeloma. Life 2024, 14, 461. https://doi.org/10.3390/life14040461

AMA Style

Radhakrishnan V, Golla U, Kudva AK. Role of Immune Cells and Immunotherapy in Multiple Myeloma. Life. 2024; 14(4):461. https://doi.org/10.3390/life14040461

Chicago/Turabian Style

Radhakrishnan, Vijay, Upendarrao Golla, and Avinash Kundadka Kudva. 2024. "Role of Immune Cells and Immunotherapy in Multiple Myeloma" Life 14, no. 4: 461. https://doi.org/10.3390/life14040461

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