Next Article in Journal
Live Biotherapeutic Lactococcus lactis GEN3013 Enhances Antitumor Efficacy of Cancer Treatment via Modulation of Cancer Progression and Immune System
Previous Article in Journal
BMS-986158, a Small Molecule Inhibitor of the Bromodomain and Extraterminal Domain Proteins, in Patients with Selected Advanced Solid Tumors: Results from a Phase 1/2a Trial
Previous Article in Special Issue
Recent Advances on the Pathobiology and Treatment of Multiple Myeloma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multiple Myeloma Therapy: Emerging Trends and Challenges

1
Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
2
Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
3
Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
4
Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
5
Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(17), 4082; https://doi.org/10.3390/cancers14174082
Submission received: 15 July 2022 / Revised: 16 August 2022 / Accepted: 18 August 2022 / Published: 23 August 2022
(This article belongs to the Special Issue Recent Advances on the Pathobiology and Treatment of Multiple Myeloma)

Abstract

:

Simple Summary

Multiple myeloma (MM) is a frequent hematological malignancy characterized by the uncontrolled growth of clonal plasma cells, primarily in the bone marrow. Over the past years, novel therapies have been discovered and introduced into clinical practice that have dramatically changed the treatment landscape of MM. Despite the tremendous advances, MM remains incurable, with poor outcomes particularly in patients with relapsed/refractory disease, emphasizing the need for new therapeutic approaches. In this review we discuss the current treatment paradigm of multiple myeloma and focus on promising future approaches.

Abstract

Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.

1. Introduction

Multiple myeloma (MM) is the second most common hematologic malignancy, characterized by the uncontrolled growth of clonal plasma cells (PCs). These cells are hyperproliferative differentiated B-lymphocytes capable of secreting a variety of immunoglobulins (Ig) [1] with the most common being IgG, IgA, and, to a lesser extent, IgD. The abnormal PCs typically grow in the bone marrow with only a small fraction of patients presenting with extramedullary disease at the time of diagnosis or developing extramedullary disease later in the disease course [2,3]. The excessive monoclonal Ig secretion from the clonal PCs leads to organ damage with the most frequent clinical symptomatology including anemia, renal failure, hypercalcemia, and lytic bone lesions [4].
Over the past few decades, rigorous pre-clinical and clinical research has led to the discovery of novel therapies that have dramatically changed the treatment landscape of MM in the frontline as well as the relapsed/refractory setting (Figure 1). The introduction of these agents has translated into prolonged progression-free periods as well as overall survival with significantly less toxicity and improved quality of life [5,6]. However, despite these tremendous advances, MM remains largely incurable and very heterogeneous, with poor outcomes especially among patients who are resistant to multiple drug classes, emphasizing the need for a better understanding of its underlying biology and the development of more effective therapeutic strategies. We will discuss herein the current treatment paradigm of multiple myeloma and focus on promising future approaches.

2. Evolution and Molecular Basis of Multiple Myeloma

Recently, there has been an increasing interest in unraveling the underlying genomics of MM in an effort to better understand the clinical heterogeneity of this complex disease, characterize its precursor states, and elucidate the significant cytogenetic and molecular events leading to active myeloma. Understanding the molecular basis of myeloma is also believed to improve risk stratification, prognostication, and prediction of response to available therapies, altogether advancing precision medicine.
At present, it is well known that MM is a progressive disorder with distinct developmental stages. Monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM) and active MM form the disease continuum characterized by progressive clonal evolution [7,8]. The gradual accumulation of cytogenetic abnormalities with translocations (such as IgH translocation) and/or gains and/or losses of chromosomes (such as loss of 1 p, 17 p, monosomies 13, 14, 17 or 1 q gain), as well as genetic mutations and epigenetic alterations in PCs over time, lead to disease progression ultimately resulting in the malignant phenotype [9,10,11,12].
Within the expanding tumor, there are genetically distinct subclones generating fluctuating intertumoral heterogeneity with particular subclones gaining selective advantage and dominating. Comparison of whole-genome sequencing data (WGS) of patients at precursor stages and with active MM showed that the sub-clonal heterogeneity is present in the early stages of MGUS and persists throughout the disease course, suggesting the presence of a branching clonal evolution pattern [13,14]. Microenvironment may also play a role in the transition from precursor disease to symptomatic myeloma [15].
MM is nearly always proceeded by MGUS; however, only ~10% of patients with MM have a prior MGUS diagnosis. This may be related to the fact that MGUS is asymptomatic and most cases are incidentally found on routine checks. The annual risk of MGUS progression to MM is around 1%; however, for high-risk MGUS, the risk almost doubles at 2% [16]. Recent findings suggest that the likelihood of conversion to a higher risk MGUS state (defined by an increase in serum biomarkers, Figure 2) can change over time, supporting continued follow up and risk assessment in MGUS patients [17].
Smoldering myeloma is a very heterogenous intermediate stage, between MGUS and MM, with variable risk for progression. Many cases are shown to have a low mutational burden and called MGUS-like; however, a high mutational burden, similar to symptomatic MM, has also been observed [7]. A phase 2 trial of carfilzomib, lenalidomide, and dexamethasone in high-risk SMM performed whole-exome sequencing (WES) of 18 SMM patients. When compared to newly diagnosed MM (NDMM), SMM patients had a significantly lower frequency of mutations in significant myeloma and NF-κB pathway genes [18].
To explore the genomic changes at the level of active disease, a study from Chapman, et al. evaluated 38 patients with NDMM, using either WGS (23 patients) or WES (16 patients). They found frequent mutations in genes known to be involved in myelomagenesis (NRAS, KRAS, TP53, CCND1), protein translation (DIS3, FAM46C), histone methylation, and the NF-κB pathway [19]. A larger study from the same group using the next-generation sequencing (NGS) technique for WGS (177 patients) or WES (26 patients), adding valuable information regarding the clonal architecture of MM and reporting the presence of different sub-clonal populations frequently harboring multiple mutations inside the same pathway (e.g., KRAS, NRAS) within the same patient [20].
WES of 463 patient with NDMM of the Myeloma XI trial also identified significant alterations in 15 genes including KRAS, NRAS, TP53, DIS3, and IRF4. Mutations in the RAS and NF-κB pathways were common at 43% and 17% respectively; however, they were not associated with the prognosis [10]. On the contrary, mutations in CCND1 and the DNA repair pathway (TP53, ATM, ATR, and ZNFHX4), chromosome 1q amplification, and MYC translocations were associated with inferior progression-free and overall survival outcomes [10].
NGS for WES had also been utilized in patients with advanced disease, confirming the high variability of genomic architecture in MM and additionally describing a significant rate of inactivating mutations in SP140 and LTB tumor suppressor genes. Increased risk of relapse was noted with SP140 mutations [21]. It was further shown that heterogeneity of the sub-clonal structure and mutational reserve of MM is generated by at least two mutational processes, which can independently change/grow over time, demonstrating distinct branching clonal evolution. Higher numbers of mutations have been associated worse survival outcomes, regardless of how mutations are generated or distributed across subclones [21].

3. Factors Influencing Treatment Strategy and Current Challenges

Several patient-related, disease-related, and treatment-related factors should be considered when selecting therapy for MM. The most important patient-related factors include age, frailty, and performance status. Disease-related parameters include the nature of the disease, for example risk status, and extent of organ damage. Treatment-related factors refer to drug availability and drug adverse events; in the case of relapsed/refractory disease the number and type of prior treatments, as well as depth and duration of previous responses, should also be taken into consideration.
Risk stratification at diagnosis is important for treatment selection. There are several scoring systems that risk stratify myeloma and its predecessor states (Figure 2). The simplest and most commonly applied system for MM is the International Staging System (ISS), which uses two parameters: the serum β-2 microglobulin and serum albumin at diagnosis [22]. The revised version referred to as R-ISS additionally incorporates high-risk chromosomal abnormalities (del (17 p), t (4;14), and t (4;16)) and serum lactate dehydrogenase [23]. Recent data for the Myeloma Genome Project that utilized NGS for WGS and WES reported that the biallelic inactivation of the TP53 gene or amplification (≥4 copies) of CKS1B (1q chromosome) in patients with ISS III were associated with poor prognosis; however, these molecular tests are not routinely done [24].
In the era of NGS utilization, several gene expression profiles (GEP) have attempted to risk stratify patients with NDMM. However, so far only the EMC-92 and UAMS GEP70 have been validated for their prognostic utility and developed into clinical tests [25,26]. A recent study by Shah et al., applied the EMC-92 GEP to the 329 NDMM transplant-eligible patients of the Myeloma XI trial and found that the EMC-92 was able to precisely identify patients with high-risk MM [27]. Moreover, they showed an independent association of EMC-92 and high-risk cytogenetic aberrations with clinical outcomes. Despite this encouraging ability for accurate risk stratification, molecular signatures are still in the embryonic stages with regards to application to clinical practice.
With respect to regimen selection, if patients are fit enough, three-drug regimens are always preferred over two-drug regimens, as they have consistently shown better responses and survival outcomes. However, frail patients may only be able to tolerate duplet therapy. Despite implementing multimodal approaches to treat MM, the major challenge remains that the vast majority of patient eventually relapse and become refractory to multiple drug classes. Additionally, patients require continuous treatment throughout the disease course, which can negatively affect their quality of life due to potential therapy-related side effects. Some patients, due to significant comorbidities, may not be candidates for specific systemic agents or autologous transplantation, which can significantly limit the suitable pool of modalities that can be utilized against their disease. Moreover, despite recent introduction of several novel agents into routine practice, there are no reliable markers that can successfully predict the response to specific drug classes, restricting our ability to select a more personalized therapeutic strategy.

4. Traditional Therapies

4.1. Alkylating Agents

Historically, alkylating agents have been used for the treatment of MM. Most alkylators work similarly by directly damaging DNA. In detail, they cause crosslinking and subsequent DNA strand breaks. This leads to abnormal base pairing, inhibiting cell division and resulting in apoptosis [28].
Melphalan was the first alkylator and first drug ever found to be effective against MM. Melphalan along with prednisone demonstrated a significant benefit and was eventually established as the standard of care in 1969 [29]. Given the introduction of novel agents over the past 20 years, melphalan-based regimens are rarely used in the US now, with two exceptions only: (1) high-dose melphalan is still the preferred conditioning regimen prior to autologous hematopoetic cell transplant (AHCT) and (2) it is used in combination with bortezomib, dexamethasone, cisplatin, doxorubicin, cyclophosphamide, and etoposide (VTD-PACE) for very aggressive disease.
Cyclophosphamide is another popular alkylator, currently used in combination with other standard-of-care agents. Cyclophosphamide has been shown to additionally have a significant immunomodulatory effect by activating NK cells, T helper cells, and macrophages [29]. Melflufen is a newer peptide–drug conjugate that targets aminopeptidases and rapidly releases the alkylator payload into the MM cells. [30] This irreversibly damages DNA and results in cell death via a p53-independent mechanism. It was recently granted accelerated approval by the FDA; however, shortly thereafter, it was withdrawn due to inferior survival compared to standard of care. Further trials are required to explore its potential.

4.2. Immunomodulatory Drugs

Immunomodulatory drugs (IMiD) have been an important component of the backbone of therapy for MM. The first-generation IMiD Thalidomide was introduced into clinical practice in the late 1990’s, followed by approval of the next-generation IMiD Lenalidomide and Pomalidomide. These were observed to be more effective with a higher therapeutic index, justifying their current widespread use in routine clinical practice [31].
IMiD hold a pleiotropic mechanism of action, including direct cytotoxicity via cell cycle arrest, induction of apoptosis as well as inhibition of cell adhesion molecules, and expression of early growth genes [31]. The cytotoxicity of IMiD is mediated by their binding to cereblon, an adapter protein linked to E3 ubiquitin protein ligase complex CRL4. This interaction induces ubiquitination and degradation of the transcription factors Ikaros and Aiolos, which are important for lymphocyte development and proliferation, ultimately resulting in MM cell apoptosis [32,33].
In addition, IMiD have immunostimulatory activity leading to increased CD4+ and CD8+ T-cell co-stimulation and secretion of IL-2 and IFN-γ, which can lead to activation of the Natural Killer (NK) cell compartment and antibody-dependent cellular cytotoxicity (ADCC) [34,35,36,37]. NK cell activation by IMiD has also been shown to occur via activation of Zap-70 and degradation of both Ikaros and Aiolos [38]. Of note, Ikaros and Aiolos suppress IL2 production in immune cells, thus IMiD increase the production of IL2 leading to T and NK cell proliferation and suppression of regulatory T cell (Tregs) function [38,39]. IMiD can also boost the immune response by enhancing antigen presentation and increasing the activity of dendritic cells (DCs) [31,40]. They also interfere with the tumor micro-environment by disrupting angiogenesis, adhesion molecules, and the stromal-MM cell interaction, altering cytokine and growth factor secretion and inhibiting osteoclastogenic proliferation [31].
Multiple preclinical and clinical trials have confirmed the efficacy of IMiD. They are currently approved as monotherapy or in several combinations with one or two or three other anti-myeloma agents in the frontline, maintenance, and the relapsed settings (Table 1).
A next generation of immunomodulators, called cereblon E3 ligase immunomodulators (CELMoDs), is presently under clinical investigation. CELMoDs and IMiD have a similar mechanism of action but CELMoDs differ in that they bind to cereblon more avidly, thus lowering its cellular concentration more potently than IMiD [41]. This leads to a much deeper and durable depletion of Ikaros and Aiolos, which are overexpressed in MM, promoting tumor growth and proliferation.
The CELMoD Iberdomide (also known as CC-220) has shown enhanced tumoricidal and immune stimulatory effects in preclinical studies [42]. The first phase 1/2, open-label, dose-escalation study of Iberdomide in combination with dexamethasone in heavily pretreated MM patients (mostly IMiD resistant) demonstrated an overall response rate of 32% and good tolerability [43]. Patients who were refractory to other IMiD had a similar ORR of 35%. Iberdomide was then combined with daratumumab, or bortezomib or carfilzomib (NCT02773030), with good responses and a favorable safety profile, with very few grade 3–4 adverse events. ORR were 46%, 56%, and 50% for the three Iberdomide-based combinations, respectively [44]. It appears that Iberdomide might be able to overcome resistance to both lenalidomide and pomalidomide, and this is awaited to be confirmed in future phase 3 randomized trials.
Another novel CELMoD that works similarly to Iberdomide is CC-92480, which has demonstrated potent synergy and safety when combined with dexamethasone in the phase 1 CC-92480 MM-01 trial (NCT03374085) that included heavily pretreated patients with relapsed/refractory MM (RRMM) [45,46]. The positive outcomes resulted in expansion of this study to CC-92480-MM-002 (NCT03989414) that currently evaluates the combination of CC-92480 with other agents including proteasome inhibitors and the anti-CD38 monoclonal antibody. Preliminary results are encouraging. Avadomide (CC-122), another cereblon-modulating agent with potent biological activities, was tested in a phase 1 trial and demonstrated acceptable safety and favorable pharmacokinetics (NCT01421524) [47]. CFT7455, a next-generation Ikaros/Aiolos degrader, is currently being tested in a phase 1 trial with dose escalation and dose expansion phases (NCT04756726) [48].

4.3. Proteasome Inhibitors

Proteasome Inhibitors (PI) have been an instrumental therapeutic class in MM. They primarily act by disrupting the natural degradation of intracellular proteins via inhibition of the proteasome complex inducing direct apoptosis. PI also downregulate the NF-κB signaling pathway and block the activation of anti-apoptotic genes, leading to cell death [49,50]. Additionally, PI have other properties including suppression of adhesion molecules and angiogenesis [51]. There is recent evidence that PI can have an immune stimulatory effect including activation of NK cells and DCs, which ultimately results in immune-mediated destruction of the malignant PCs [52,53,54].
At present, there are three PI approved for the treatment of MM—bortezomib (V), carfilzomib (K), and ixazomib [55]. Bortezomib, a first-generation PI, has a slow reversible effect on the proteasome, whereas carfilzomib, a second-generation PI, binds the proteasome irreversibly and more selectively [55,56]. Additionally, V inhibits both chymotrypsin-like and caspase-like activities of the proteasome, whereas K is more selective for the chymotrypsin-like active site within the proteasome [56]. Therefore, K holds greater specificity and potency and a different safety profile compared to V. Ixazomib is the only orally bioavailable PI, with properties similar to V, but with a more begnin neutotoxicity adverse event profile [57,58]. PI are typically used in doublet, triplet, or quadruplet combinations with other drug classes such as IMiD and monoclonal antibodies for newly diagnosed as well as RRMM (Table 1).
Other, newer PI, still under investigation in the preclinical or phase 1/2 level, are marizomib, oprozomib, and delanzomib. Marizomib is a potent irreversible inhibitor of all three core enzyme activities of the 20 S proteasome [59]. It is currently studied as a single agent or in combination with pomalidomide and dexamethasone in previously treated RRMM patients. Oprozomib is an orally bioavailable PI that primarily inhibits the chymotrypsin-like activity of the proteasome and works similarly to carfilzomib [60,61]. It is being studied as monotherapy or along with IMiD, steroids, or alkylating agents. Two other oral PI, LC53-0110 and LC53-0151, have only been studied in mice xenografts, where they have demonstrated remarkable potency [62]; however, they have not been tested in humans yet.

5. Immunotherapy

5.1. Immune System Dysreguation

A hallmark of the underlying biology of MM is the immune system dysfunction, which is caused by various mechanisms and is believed to play a central role in the pathogenesis of the disease by promoting clonal cell proliferation via immune escape and contributing to drug resistance [63,64]. Loss of tumor antigenicity via impaired expression or alterations of tumor antigens on the surface of MM cells and upregulation of inhibitory surface ligands can lead to tumor escape from immune surveillance, along with defects in antigen processing/presentation [65,66,67,68,69,70,71,72]. This is supported by the robust T-cell response to MM antigens in bone marrow samples of patients with MGUS but absence of this phenomenon in the bone marrow of patients with active MM, despite similar clonal PCs populations [73,74].
The dysregulation further involves the tumor microenvironment, including alterations in the T and NK compartments, with upregulation of inhibitory molecules/ligands, resulting in an immunosuppressive milieu [75,76,77,78,79,80]. The increased recruitment of immunosuppressive cells such as Tregs, regulatory B cells (Bregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), along with the simultaneous reduction in cytotoxic T lymphocyte (CTLs) and defective function of antigen-presenting DCs, leads to decreased humoral and cytotoxic immunity [76,80,81,82,83]. Several novel agents that have been developed over the past decade are subsumed under the umbrella of immunotherapy and target different aspects of the immune system to eradicate MM cells. These agents include monoclonal antibodies, immune checkpoint inhibitors, bispecific antibodies, genetically engineered immune cells, and peptide vaccines.

5.2. Naked Monocloal Antibodies

Naked monoclonal antibodies (mAb) target antigens primarily expressed on the surface of PCs and lead to cell death via several different mechanisms. Currently, there are multiple antigens studied as potential targets, with the most important being CD38 and signaling-lymphocyte-activating molecule family-7 (SLAMF7), against which mAb have been developed are broadly used in clinical practice.

5.2.1. Anti-CD38 mAb

Daratumumab is a humanized mAb that targets a unique epitope on the CD38 transmembrane glycoprotein, predominately located at the surface of PCs [84]. CD38 is a multifunctional ectoenzyme that modulates NAD+ degradation and plays a critical role in the synthesis of cyclic-ADP-ribose, properties that contribute to cell survival via intracellular calcium mobilization and homeostasis [84,85,86,87,88].
The binding of daratumumab to its target induces cell death through various immune effector mechanisms. These mechanisms include complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis, and apoptosis via crosslinking [89,90,91]. Moreover, inhibiting the ectoenzymatic function of CD38 can lead to the direct apoptosis of neoplastic cells [88]. Another mechanism of daratumumab action is linked to its immunomodulatory activity, due to the presence of CD38 on various immune cells such as Tregs, Bregs, and MDSC [92].
There is significant clinical evidence for the high efficacy of daratumumab as a single agent or in combination with other drug classes [93,94,95,96] (Table 1). Pre-clinical studies have demonstrated its synergistic action with IMiD, which upregulate the expression of CD38 on PCs and therefore sensitize them to anti-CD38 mAb [97].
Isatuximab is a chimeric anti-CD38 mAb that works similarly to daratumumab [89]. Isatuximab binds to a different epitope of the CD38 surface receptor, alleviating the immunosuppressive microenvironment of the bone marrow niche in MM [98,99]. It is currently approved in combination with the duplets of pomalidomide–dexamethasone and carfilzomib–dexamethasone for RRMM (Table 1), with ongoing clinical trials testing various other combinations in the relapsed and frontline settings [100,101].

5.2.2. Anti-SLAMF7 mAb

Signaling-lymphocyte-activating molecule family-7 (SLAMF7) is a receptor that is highly expressed on the surface of PCs but also other leukocytes, especially NK cells [102,103,104,105]. It has immunomodulatory properties and mediates the interaction of MM cells among each other and also with tumor microenvironment stroma, activating several downstream pathways that promote malignant PCs proliferation. [106,107].
Elotuzumab is an anti-SLAM7 humanized mAb whose Fc portion binds to the CD16 on the surface of NK cells, constituting a bridge between MM and NK cells [108,109]. This attachment leads to the activation of NK cells, tagging MM cells for destruction in an ADCC-dependent manner [106]. There is preclinical evidence that augmenting elotuzumab NK cell-mediated ADCC can enhance its anti-tumor effect. This can be achieved by the blockade of the killer immunoglobulin-like checkpoint receptors (KIR) with the experimental mAb lirilumab, or by activating the CD137 receptor with the mAb Urelumab. However, these combinations are yet to be studied in the clinical context [110,111]. Elotuzumab can also directly activate NK cells by binding to SLAMF7 on their surface, hence facilitating the downstream activation of EAT2, leading to enhanced phosphorylation of ERK [112]. Given that the activity of elotuzumab is strongly dependent on the activity of NK cells, dysfunction in the NK compartment can lead to decreased efficacy. Moreover, elotuzumab can interfere with the tumor microenvironment by disrupting the adhesion of MM cells to the bone marrow stromal cells [107].
Initial experimental studies demonstrated that elotuzumab is more effective when combined with traditional antimyeloma agents such as lenalidomide and bortezomib [113,114]. This was further confirmed in clinical trials which found elotuzumab to have modest activity as monotherapy [115], and better activity when combined with lenalidomide or pomalidomide [116,117]. Elotuzumab is currently approved with pomalidomide/dexamethasone or lenalidomide/dexamethasone for relapsed/refractory disease (Table 1). Elotuzumab has also been studied in combination with bortezomib (NCT01478048) with encouraging results. A phase 3 trial exploring the addition of elotuzumab to the triplet carfilzomib/lenalidomide/dexamethasone in the upfront and post-transplant maintenance setting is ongoing (NCT03948035).

5.3. Immune Checkpoint Inhibitors

Immune checkpoints are inhibitory receptors on the surface of T cells that mediate immune tolerance to self-antigens, by suppressing the T cell compartment when activated during the antigen presenting process [118,119]. There is evidence that the cytotoxic tumor lymphocyte antigen 4 (CTLA-4) and programmed cell death-1 (PD-1) immune checkpoints are highly expressed on the surface of T, B, and NK cells in the bone marrow of patients with MM [120,121]. The binding of these receptors to their ligands on antigen presenting cells (APC) and/or tumor cells leads to suppression of cytotoxic T cells and upregulation of Tregs, thus inhibiting the immune response, favoring cancer cell growth via immune escape [119,120,122]. Experiments have shown that PD-L1, the major ligand of the PD-1 receptor, can be upregulated on malignant PCs [122]. MM cells with high PD-L1 expression appear to be more proliferative and resistant to therapy, indicating increased aggressiveness [123]. T cell immunoreceptor with Ig and ITIM domains (TIGIT) and lymphocyte activation gene-3 (LAG-3 or CD223) are other immune checkpoints on the surface of T cells involved in T cell regulation by activating Tregs and inhibiting cytotoxic T cells [124,125,126,127]. Increased expression of LAG3 on T cells in the bone marrows of MM patients is associated with sustained T cell stimulation leading to T cell exhaustion, which can potentially contribute to immune escape [128].
Immune checkpoint inhibitors (ICI) are a distinct category of “naked” mAb targeting molecules that constitute immune checkpoints. The ICI mainly evaluated in MM are the anti-PD-L1 mAb, which block the binding of PD-L1 to its PD-1 receptor. PD-1/PD-L1 blockade alone was not efficacious in phase 1 studies [128,129]; however, combination approaches with different drug classes such as IMiD and anti-CD38 mAb appeared promising. In preclinical studies, IMiD reduced the expression of PD-1 receptors on T cell surfaces and also down-regulated PD-L1 on MM cells, supporting a potential synergetic effect with PD-L1 inhibitors [130]. In vivo studies have also shown that long exposure to PD-1 blockade enhances the anti-CD38 ADCC, suggesting a potential clinical benefit to combining anti-PD-L1 ICI with anti-CD-38 mAb [131].
Clinically, several studies have evaluated PD-1 blockade using pembrolizumab or nivolumab with IMiD such as lenalidomide and pomalidomide; however, they failed to demonstrate improvement in disease response. Interestingly, a combination of pembrolizumab with lenalidomide was associated with high rates of toxicity and increased risk of death; thus, the FDA put a hold on studies investigating combinations of anti-PD-L1 ICI with IMiD [132,133,134]. Anti-PD-L1 inhibitors have also been used in combination with daratumumab without safety warnings but no clinical benefit so far [135,136,137,138]. Nivolumab is currently being tested in combination with carfilzomib and pelareorep in a phase 1 trial (NCT03605719). More recent phase 1 and 2 trials are examining the efficacy and safety of anti-LAG 3 (BMS-986207) and anti-TIGIT (BMS-986207 or COM902) ICI alone or combined with other agents (NCT04354246, NCT04150965). To date, the use of ICI alone or in combination with traditional anti-myeloma agents has not proven efficacious in the clinical setting and is currently not recommended.

5.4. Antibody Drug Conjugates

A novel type of therapy that has recently been investigated in the clinical setting is the antibody drug conjugates (ADC). ADC are mAb against a specific tumor target on the surface of malignant cells that carry a small cytotoxic agent (payload), such as microtubule inhibitors and agents damaging DNA, utilizing a cleavable or non-cleavable linker [139,140]. When it reaches its target, the ADC is internalized with eventual release of the payload into the cytoplasm of malignant PCs, leading to cell death [140] (Figure 3). Cleavable linkers are degradated by enzymes in the cytoplasm of the malignant cells, whereas non-cleavable linkers require processing and degradation of the mAb complex into the lysosomes in order to release the toxic payload [140]. The target of ADC should ideally be a molecule highly expressed on the surface of malignant PCs with very low or no expression on other cell types, including hematopoeitic cells, to avoid systemic toxicity [141]. ADC can also exert their effects via ADCC, ADCP, or CDC [142,143].

5.4.1. B Cell Maturation Antigen (BCMA)

The most well-studied ADC target to date is BCMA, which is a member of the tumor necrosis factor receptor family exclusively expressed on plasma cells and late mature B cells [144]. Activation of BCMA by its ligands, B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL), is required for the survival of PCs [145,146]. At present, there are several different ADCs under development that target the BCMA carrying different types of linkers and payloads.
The anti-BCMA ADC belantamab mafodotin (GSK2857916) is a humanized IgG1 anti-BCMA mAb carrying a non-cleavable linker to the tubulin polymerization inhibitor monomethyl aurastatin-F (mcMMAF) [143]. Apart from the classic mechanism of cell death after the endoplasmic release of mcMMAF, belantamab mafodotin can also induce cell lysis via effector-cell-mediated ADCC, with the defucosylated Fc region of the mAb helping to facilitate this interaction [143,147]. Malignant cell lysis releases tumor antigens, locally triggering a robust immune response against myeloma cells. [146,147]. The clinical safety and efficacy of belantamab mafodotin monotherapy in heavily pretreated MM patients were confirmed in the DREAMM-1 and DREAMM-2 clinical trials [148,149,150]. At present, belantamab mafodotin is being studied in combination with other anti-myeloma agents in phase 2 and 3 clinical trials (NCT03715478, NCT03544281, NCT04246047 and NCT04484623). To date, it is the only ADC approved by the FDA, as a single agent, for the treatment of MM patients who have received at least four prior therapies including an anti-CD38 mAb, a PI, and an IMiD [149,151,152].
Other anti-BCMA ADC include AMG224, MEDI2228, CC-99712, and HDP-101. AMG224 is a mAb conjugated with antitubulin maytainsinoid using a noncleavable linker, currently studied as monotherapy in a phase 1 trial in heavily pretreated MM patients (NCT02561962) [153,154]. MEDI2228 is another BCMA-targeted ADC conjugated with a DNA cross-linking pyrrolobenzodiazepine using a protease cleavable linker [155]. After binding to BCMA, the complex is internalized and cleaved in the lysosomes, releasing the toxic payload, which causes DNA damage and apoptosis [155]. In preclinical studies, it demonstrated synergism with bortezomib [156,157]. A phase 1 study assessed its safety as a single agent in the RRMM setting, reporting encouraging clinical efficacy and a manageable safety profile (NCT03489525) [158]. The newer anti-BCMA ADCs, CC-99712 and HDP-101, are also in early phase studies (NCT04036461, NCT04879043).

5.4.2. Other ADC Targets

CD38 is a glycoprotein on the membrane of PCs targeted by the “naked” mAb daratumumab and isatuximab, with robust clinical efficacy [85]. MT-0169 or TAK-169 is an investigational ADC that similarly targets the CD38 receptor; however, it is conjugated with a ribosome-inactivating Shiga-like engineered toxin called (SLTA) [159]. In vivo pre-clinical data have been encouraging, leading to a phase 1 (NCT04017130) study. TAK-573 is another anti-CD38 IgG4 ADC genetically fused to two attenuated interferon alpha-2b (IFNα2b) molecules. Notably, this agent binds to a location of the CD38 receptor that is different from the binding sites of daratumumab and isatuximab [160]. Given promising outcomes in xenograft models, there is an ongoing a phase 1/2 trial testing TAK-573 in combination with dexamethasone (NCT03215030).
CD46 is a transmembrane protein that is highly expressed in MM cells that is involved in complement inhibition [161]. An anti-CD46 ADC is fused with saporin and subsequently to monomethylauristatin F (MMAF) [161]. Preclinical data have demonstrated anti-myeloma activity, although testing in humans has not been initiated yet. FOR46, another anti-CD46 ADC with an undisclosed payload, is currently being tested in a phase 1 trial for RRMM (NCT03650491).
CD74 is a glycoprotein found on the surface of the majority of PCs and is involved in the antigen presentation process by the major histocompatibility complex class II [162,163]. STRO-001 is a human anti-CD74 IgG fused with a potent maytansinoid warhead to two specific sites [164]. This agent is being studied in an ongoing phase 1 trial (NCT03424603) with encouraging preliminary results [165].

5.5. Bispecific Antibodies

Bispecific antibodies (bsAbs) are mAb designed to bind to a target on the surface of the malignant myeloma cells and effector cells (T or NK cells), creating an immunologic bridge leading to the destruction of the tumor cell by the activated effector cell [166] (Figure 3). There are several bsAbs currently being tested in the preclinical and clinical settings. The most popular antigenic targets on PCs include BCMA, CD38, GPRC5D, and FcRH5 [167]. GPRC5D is a G-protein–coupled receptor with unclear function that is highly expressed on the surface of myeloma cells [168]. FcRH5 belongs to the immunoglobulin superfamily and is located only on the surface of B cells with increasing expression on myeloma cells [169,170]. At present, all bsAbs in clinical trials target the CD3 on the surface of T cells [167]. However, in a preclinical level, bispecific NK-cell engagers are also under investigation with good anti-myeloma activity [171,172,173].

5.5.1. Bispecific T Cell Engagers

There are several bispecific antibodies targeting BCMA and CD3 on T cells (BCMAxCD3) that induce T cell activation against the tumor and lead to lysis of MM cells [174]. The main ones currently being studied in the clinical setting include: AMG420, AMG701, CC-93269, elranatamab (PF-06863135), REGN5458, REGN5459, teclistamab (JNJ-64007957), and TNB-383B [175,176,177,178] (Table 2). Bispecific T cell engagers targeting GPRC5D on tumor cells (GPRC5DxCD3) include talquetamab (JNJ-64407564) [179,180,181], whereas the ones targeting CD38 (CD38xCD3) and FcRH5 (FcRH5xCD3) on PCs are TNB-383B and cevostamab, respectively [182,183]. Development of trispecific antibodies or trispecific T cell engagers is a step forward for bsAbs, by adding a T cell costimulatory signal such as CD28 or dual MM targeting in an attempt to overcome the possibility of T cell anergy, which can lead to suboptimal responses [184,185]. Preclinical testing of a trispecific antibody targeting the CD38, CD3, and CD28 on T cells in mice xenografts, has demonstrated a very potent antimyeloma effect [184]. However, no trispecific antibody has entered clinical testing in humans yet. At present, several bispecific and trispecific antibodies activating the T cell compartment are under rigorous investigation and awaiting advancement to the clinic.

5.5.2. Bispecific NK Cell Engagers

Bispecific NK cell engagers are bispecific antibodies that redirect the NK cell compartment against the tumor cells via binding to a receptor on the NK cell surface such as CD16A, NKp30, or NKG2d and to a molecule on the surface of the malignant PCs, such as the BCMA or SLAM7 [171,172,173]. Several agents are under development with the following combinations: BCMAxCD16a (AFM26, RO7297089), BCMAxNKp30 (CTX-8573), and SLAM7xNKG2D (CS1-NKG2D) [171,172,173,186,187]. A trispecific NK cell engager targeting the BCMA, as well as the CD20 co-stimulatory molecule and CD16a on NK cells, is also under preclinical stages of development [188]. Clinical trials of this agent class are eagerly awaited.

5.6. Chimeric Antigen Receptor (CAR) T Cell Therapy

Chimeric antigen receptors (CAR) are synthetic transmembrane receptors that are designed to selectively recognize specific antigens on the surface of target cells [189,190]. The extracellular antigen recognition domain typically consists of a single-chain variable fragment (scFv), whereas the intracellular activation domain is typically derived from the CD3ζ chain that subsequently induces T cell activation upon antigen binding [191,192,193] (Figure 3). First-generation CAR lacked a costimulatory domain, resulting in only moderate responses [194]. However, the next-generation CAR included co-stimulatory signaling endodomains, such as CD28, CD137 (4-1BB), or inducible T cell co-stimulator (ICOS), in an attempt to mimic the co-stimulation occurring during physiological T cell activation via TCR recognition by APC, with subsequent improvement in T cell responses [195].
The CAR T cell production starts with collection of T cells from patients and continues with the transfer of the gene encoding the CAR construct into the genome of these T cells using a viral vector [196,197]. The CAR gene is subsequently transcribed and expressed as a surface receptor [198,199]. CAR T cell manufacturing occurs ex vivo and takes 4 weeks on average [198,199]. CAR T cell therapy is typically given as a single infusion after the administration of lymphodepleting chemotherapy, which facilitates the proliferation and activity of CAR T cells [200,201].
The choice of target antigen is critical, as it needs to be uniformly expressed on malignant cells with minimal expression on other hematopoietic cells and tissues [198]. BCMA was the first antigen to be targeted in CAR T cell therapy clinical trials [145]. Idecabtagene vicleucel (Ide-cel) was the first CAR T product officially approved for heavily pretreated MM patients, followed by ciltacabtagene autoleucel (cilta-cel), both of which target BCMA on the surface of myeloma cells [202,203]. Ide-cel is composed of a mouse scFv (11D5-3) targeting domain, a 4-1BB (CD137) co-stimulatory domain, and a CD3ζ T-cell activation domain, and uses a lentivirus vector for CAR introduction into the genome of T cells [204,205]. On the other hand, cilta-cel is composed of two llama-derived variable heavy-chain-only (non-scFv) antigen recognition domains targeting two distinct regions of BCMA, a 4-1BB (CD137) co-stimulatory domain, and a CD3ζ T cell activation domain, and uses a lentivirus vector similar to ide-cel [206,207].
Despite the associated high responses, not all patients have durable responses after CAR T cell therapy [208], which is related to several tumor- and CAR-T-cell-construct-related factors [208]. Given that most CAR T cell products target BCMA, there is evidence suggesting that low baseline BCMA expression levels on tumor cells negatively impacts the efficacy of CAR T cells [209]. Additionally, myeloma cells can shed BCMA, leading to lower surface concentration and circulation of soluble BCMA (sBCMA). sBCMA binds to CAR T cells, blocking their interactions with BCMA on the surface of malignant cells, resulting in the decreased efficacy of CAR T cells, as shown in preclinical studies [210,211]. One mechanism that could explain antigenic loss is acquired biallelic BCMA deletion, resulting in decreased BCMA expression [212,213]. High tumor load also appears to negatively affect the efficacy of CAR T cell therapy, perhaps due to CAR T cell exhaustion [202,214]. High expression of immune checkpoints on the surface on myeloma and CAR T cells can also attenuate CAR T cell activity [193,215,216]. CAR T cells typically induce malignant cell death via the release of toxic granules containing perforin and serine proteases, and the induction of apoptosis via receptor cross-linking. It has been described that, in cases of treatment resistance, tumor cells were found to overexpress several antiapoptotic molecules including serine protease inhibitors or other proteins interfering with crosslinking [217,218].
The quality and composition of T cells in the leukapheresis product can also influence the outcomes of CAR T cell therapy. A high frequency of less-differentiated early memory T cells [219,220,221] and a high CD4/CD8 T cell ratio in the apheresis collection [201,221], which is typically seen in patients early in their disease course [222], leads to higher CAR T cell proliferation, expansion, and persistence and subsequently higher response rates [223,224]. On the contrary, multiple prior therapies in heavily pretreated patients are believed to negatively affect the fitness and constitution of the T-cell compartment.
There are several approaches to overcome these challenges, including dual-targeted CAR T cells harboring two different CAR or one CAR with two different antigen binding domains [225]. Several preclinical studies are currently investigating simultaneous targeting of BCMA and SLAMF7, GPRC5D, or CD38, molecules uniformly expressed on MM cells [217,225,226,227]. Another idea is to manufacture CAR T cells that can secrete checkpoint inhibitory antibodies such as anti-PD1 or anti-PD-L1 or CAR T cells in which genes that express immune checkpoints are knocked down [228,229,230]. Optimizing the structure of CAR by adding a costimulatory domains is also important as it can lead to improved persistence and activity with decreased exhaustion. [214,231,232]. In an effort to collect a more balanced T cell product, which would theoretically enhance CAR T cell function, allogeneic CAR T cells generated from the T cells of healthy donors have also been manufactured and assessed in the clinical context, with promising outcomes and an acceptable side effect profile [233].

5.7. Peptite Vaccines

An attractive approach for controlling tumors is developing synthetic peptide vaccines derived from widely expressed tumor-associated antigens (TAAs), which have the ability to bind multiple MHC class I and class II alleles, thus activating T-cell-mediated tumor destruction. This method is considered safe, and theoretically can be highly potent, specific, and long lasting. [234]. One approach is to target MUC1 (mucin 1, cell surface associated), a mucin-like glycoprotein highly expressed in a variety of epithelial and hematologic tumors including MM [235,236,237]. MUC1 is made of a large soluble extracellular alpha subunit containing the tandem repeats array (TRA) and a smaller beta subunit containing the transmembrane and cytoplasmic domains. The MUC1 signal peptide (SP) domain of the MUC1 binds multiple MHC class I and class II alleles, generating a robust T cell immunity; therefore, it was felt to serve as suitable vaccine candidate. Based on this rationale, a 21mer peptide vaccine encoding the complete signal domain of MUC1 was constructed and named as the ImMucin (VXL100) vaccine. [235,236,237]. In a phase 1/2 study, 15 MM patients were enrolled and vaccinated with ImMucin; however, only 9 patients completed the vaccination course (a total of six doses) [234]. ImMucin vaccination resulted in a significant increase in the percentage of both γ-interferon-producing CD4+ and CD8+ T cells in all patients. Additionally, a 9.4-fold increase in peripheral blood mononuclear cells and a 6.8-fold increase in anti-ImMucin antibodies was noted. Disease improvement or stability persisted for 17.5–41.3 months post-vaccination. These findings suggested a potential therapeutic benefit of ImMucin in MUC1-positive tumors in MM patients.
Similarly, PVX-410 is a human leukocyte antigen (HLA)-A2-restricted multipeptide vaccine for patients with SMM [238,239]. The vaccine is composed of a unique combination of four immunogenic peptides (XBP1US184–192, XBP1SP367–375, CD138260–268, and CS1239–247) derived from specific tumor target antigens (XBP1, CD138, and CS1, respectively) highly expressed on MM cells. These peptides were found to activate the immune system in an HLA-A2-specific manner, inducing antigen-specific CTLs against HLA-A2-positive MM cells. [239]. In a phase 1/2a study, 22 patients with SMM and the presence of HLA-A2 were divided into three groups, PVX-410 (low and target dose) or lenalidomide with PVX-410 [240]. In all cohorts, the PVX-410 vaccine induced a highly effective immune response against MM cells, with expansion of the CD3+ CD8+ CTL compartment against the XBP1, CD138, and CS1 antigenic epitopes. The response was further enhanced during treatment with lenalidomide. In the target-dose cohort, 1 out of 9 patients progressed (median TTP 36 weeks), as well as 1 out of 12 in the combination cohort (median TTP no reached).
A relatively recent phase 1 trial demonstrated the role of the PD-L1 peptide (IO103) vaccine in MM patients [241]. As previously mentioned, upregulation of PD-1/PD-L1 [121,242,243] is associated with poor prognosis in patients with MM [244]. Stimulation with the IO103 peptide stimulated PD-L1-specific T cytotoxic cells against PD-L1-expressing MM cells. In this study, 10 patients with MM who were 6 months post AHCT were enrolled [241]. Patients received vaccination with IO103 up to 15 times within one year. All patients showed a peptide-specific immune response in peripheral blood mononuclear cells and in skin-infiltrating lymphocytes. Three out of ten patients had improvement of response (over 100 days post-transplant) [241].
Quian and colleagues assessed the role of Dickkopf-1 (DKK1), a protein that is highly expressed in MM cells but not in normal tissues, as a potential vaccine candidate. Their in vitro experiments showed that cytotoxic T lymphocytes were able to recognize DKK1 peptides naturally presented by MM cells in the context of HLA-A*0201 molecules. This led to the immune-mediated destruction of MM cells, hence suggesting that DKK1 could be a potentially important antigen for immunotherapy in MM [245]. Further experiments from the same group in mouse murine myeloma models showed that vaccination with DKK1-DNA not only prevented mice from developing MM, but was also therapeutic against active MM. DKK1 vaccination elicited strong DKK1- and tumor-specific CD4+ and CD8+ immune responses, providing extra evidence for targeting DKK1 in MM patients [246]. Despite these encouraging outcomes, vaccination against MM has not been adopted in the clinical setting. There is currently an ongoing pilot phase 1 study exploring the application of the DKK1 vaccine in patients with MGUS and stable or smoldering myeloma (NCT03591614).

6. Targeted Therapies and Small Molecules

6.1. Exportin Inhibitors

The nuclear pore complexes (NPC) are large cylindrical channels, composed of several copies of >30 different proteins called nucleoporins [247]. The main function of the NPC is to fuse the inner and outer nuclear membranes, enabling traffic of vital macromolecules between the nucleus and the cytoplasm, a process which is mediated by specific protein carriers, importins and exportins. [248]. Exportin-1 (XPO1) is one of the most well-characterized nuclear exporters, involved in shuttling of multiple cargo proteins such as tumor suppressor proteins, cell cycle regulators, immune response regulators, and oncogenes, as well as mRNAs, out of the nucleus and into the cytoplasm, enhancing the synthesis of oncoproteins [249,250]. Overexpression of XPO1 leads to increased transfer of tumor suppressor and regulatory proteins into the cytoplasm, which further promotes cell proliferation and halts apoptosis, overall favoring carcinogenesis. Increased XPO1 levels have been observed in a variety of malignancies including CD138+ PCs from patients with active MM and are associated with poor survival outcomes, making XPO1 an attractive molecular target for novel therapies [251]. Selective inhibitors of nuclear export (SINE) are orally bioavailable small-molecule drugs that inhibit XPO1 by attaching to the binding site of the cargo, thus disrupting the nuclear–cytoplasmic trafficking. As a result, tumor suppressor proteins and regulators eventually accumulate in the nucleus, activating the apoptotic process and subsequently causing cell death [252,253].
Selinexor is an XPO1 inhibitor that has reduced the viability of MM cells in preclinical experiments, alone or in synergism with other anti-myeloma agents. [238] In detail, selinexor causes retention of tumor suppressor proteins in the nucleus such as p53, p27, and FOXO and decreases the levels of cell cycle promoters and antiapoptotic proteins, leading to cell cycle arrest, with subsequent caspase activation and cell death [254,255,256] (Figure 4). It has also been shown to block NF-kB, which regulates osteoclast differentiation [257]. It is currently approved in combination with bortezomib/dexamethasone or dexamethasone alone in the relapsed/refractory setting, with ongoing trials investigating different combinations with other novel agents [258].

6.2. Histone Deacetylase Inhibitors

Histone deacetylase (HDAC) inhibitors act at the epigenetic level, by removing acetyl groups from mainly the histones, proteins forming the nucleosome, which play a critical role in chromatin organization [259] (Figure 4). In MM, overexpression of HDAC, especially HDAC-1, has been associated with a poor prognosis and with resistance to PI [260,261]. Panobinostat and vorinostat are pan-HDAC inhibitors leading to blockade of disposal of several proapoptotic proteins through the unfolded protein response, disrupting protein homeostasis and resulting in cell death via apoptosis [262].
Panobinostat was initially approved by the FDA; however, due to lack of confirmatory post-approval clinical studies, required as part of the accelerated approval process, it was withdrawn from the market. Other experimental HDAC inhibitors, such as quisinostat, CUDC-907, and AR-42, have also been studied in the pre- and clinical settings.

6.3. BCL2 Inhibitors

The B cell lymphoma-2 (Bcl-2) protein family consists of pro- and anti-apoptotic proteins which regulate the intrinsic pathway of apoptosis. Bcl-2 is an anti-apoptotic protein of the Bcl-2 family containing four homogeneous domains called BH1, BH2, BH3, and BH4, whereas pro-apoptotic proteins in the same family only contain the BH3 domain and are called BH3-only proteins [263]. The latter subcategory primarily works by binding to anti-apoptotic proteins, activating the BAX/BAK proteins, directly or indirectly, and inducing apoptosis [264,265]. Overexpression of the anti-apoptotic Bcl-2 has primarily been observed in the subgroup of MM patients harboring the translocation of the chromosomes 14 and 17 [266]. High levels of Bcl-2 promote cell survival and tumorigenesis and have been associated with poor outcomes and resistance to traditional anti-myeloma agents; therefore, Bcl-2 represents an attractive target for novel therapies.
Venetoclax is an orally bioavailable BH-3 mimetic that selectively inhibits Bcl-2, disrupting the anti-apoptotic pathway, thus favoring cell death in a TP-53-independent manner. (Figure 4) Venetoclax is particularly efficacious in the subset of MM patients with the translocation (11;14). These patients express high levels of Bcl-2, possibly due to increased tumoral dependence upon Bcl-2 [267]. As a result, translocation (11;14) has emerged as the first predictor of susceptibility to Bcl-2 inhibition in MM patients [268]. Venetoclax is not FDA-approved yet; however, the NCCN guidelines recommend its use in RRMM with the translocation (11;14).

6.4. Hypomethylating Agents

While hypomethylating agents have been effective for the treatment of myeloid leukemia, it seems they had limited efficacy in a phase 1b trial of 42 heavily pretreated patients with RRMM. This trial assessed the addition of Azacytidine, a DNA methylation inhibitor, to lenalidomide and dexamethasone with the purpose of overcoming refractoriness to IMiD via interfering with pathways associated with PC differentiation, apoptosis, and immune recognition. The overall response rate was 32%, with 10% achieving very good partial response, and the median PFS was 3.1 months. The levels of the azacytidine-inactivating enzyme cytidine deaminase (CDA) were measured to assess any potential correlation with treatment response, and it was found that low plasma CDA levels were associated with greater clinical benefit [269]. Currently, there is an ongoing phase II trial evaluating azacitidine in combination with daratumumab and dexamethasone in patients with RRMM who have already received daratumumab (NCT04407442).

6.5. Proteolysis-Targeting Chimera

Proteolysis-targeting chimera (PROTAC) is a class of bi-functional degrader molecules that have designed to selectively target and then degrade intractable cellular proteins via activation of the ubiquitin–proteasome system. These molecules typically consist of two ligand-binding domains, one that binds to a E3 ubiquitin ligase and another that binds a protein of interest (POI) [270]. The two domains are connected through a linker. PROTAC ultimately forms a complex between an E3 ligase and POI, which results in ubiquitination and subsequent degradation by the proteasome. The domain of PROTAC binding to an E3 ligase can be either a phthalimide derivative binding to a cereblon (CRL4 CRBN) E3 ligase (Cereblon PROTAC), or a von Hippel–Lindau (VHL) binding to VHL E3 ligase (VHL PROTAC) [270,271].
An initial PROTAC called dBET1 was constructed using thalidomide (as an E3 ligase binding domain) and JQ1 (as a POI binding domain) [272]. JQ1 is a small molecule binding to Bromodomain-Containing Protein 4 (BRD4). This PROTAC induced cereblon-dependent degradation of BRD4 and subsequent down-regulation of MYC, leading to the cytotoxicity of AML cells [273]. In vitro and in vivo pre-clinical studies in MM models using PROTAC targeting BRD4 and other BET proteins reduced the viability of MM cell lines in a time- and concentration-dependent manner and demonstrated suppressed MYC and Akt/mTOR signaling [274]. PROTAC was able to overcome resistance to PI and IMiD, and their activity was maintained in MM cells with wild-type or deleted TP53. Further studies demonstrated that BET-targeted PRTOAC was able to inhibit cell proliferation of multiple human-derived MM cell lines and fresh myeloma samples and suggested potential synergy with systemic agents including selinexor [275]. Another, newer experimental PROTAC targeting the proteasome substrate receptor hRpn13, which was found to be upregulated in MM, was tested in in vitro studies [276]. Optimization of PROTAC design for potential clinical development is eagerly awaited.

7. Emerging Approaches and Future Directions

7.1. Protein Disulfide Isomerase 1 Inhibitors

In MM, the malignant PCs exhibit very high protein synthesis burden, as evident by the abundant secretion of non-functional monoclonal antibodies and cytokines [277]. This requires re-arrangement of intramolecular disulfide bonds mediated by the protein disulfide isomerase (PDI) family in the endoplasmic reticulum (ER), an organelle that is responsible for the biosynthesis, folding, maturation, stabilization, and trafficking of transmembrane and secretory proteins [278,279]. PDIA1 is the main endoplasmic reticulum resident isoform of this multifunctional protein family, overexpression of which has been observed in a variety of cancers [280,281,282]. As previously mentioned, malignant PCs are highly dependent on proteostasis, largely taking place in the ER [283]. Disruption of the ER, which constantly functions at maximum capacity in MM cells, can cause dysregulation of the multi-level protein folding process, known as ER stress, secondary to the accumulation of unfolded or misfolded proteins, leading to activation of the unfolded protein response (UPR) and re-establishment of proteostasis and ER recovery [284]. However, in cases of severe ER stress, damage is irreversible and cell apoptosis is inevitable (Figure 5) [285].
Disruption of proteostasis has proven to be overwhelmingly efficacious in MM with the use of PI, suggesting that inhibition of proteostasis at earlier levels, alone or in combination with PI, may result in higher efficacy, especially in patients with PI-refractory disease [286]. Recently, analysis of bone marrow specimens from 690 individuals showed high expression of PDIA1 in RRMM patients that was inversely related with overall survival [287]. This observation suggested that upregulation of PDIA1 expression in patients with RRMM may confer an adaptive mechanism of the relapse/refractory state and, in part, be responsible for the disease resistance to known therapies, hence indicating that PDIA1 is a good option for targeted therapy development.
CCF642 was the first PDIA1 inhibitor constructed in the lab, which leads to irreversible lethal ER stress with subsequent MM cell apoptosis in in vitro and in vivo preclinical studies of bortezomib-resistant MM cells, without toxic effects on the healthy bone marrow hematopoietic cells [287]. However, due to poor solubility and lack of availability, it was not advanced to clinical studies. Later, a pan-PDI inhibitor, E64FC26, was tested in mice and was found to improve survival and enhance the activity of bortezomib without any adverse effects [288]. Most recently, a new, more selective PDIA1 inhibitor, CCF642-34, was developed, with improved solubility, selectivity, and potency [286].
At a molecular level, inhibition of PDIA1 with CCF642-34 inducted irreversible ER stress and cell death via PARP activation and caspase 3 cleavage, as well as increased reactive oxygen species (ROS) production, leading to upregulation of the NRF2 pathway in MM cells. Analysis of RNA NGS from MM cells demonstrated selectivity in ER stress induction, particularly via activation of UPR, ER-associated protein degradation (ERAD), and the ATF6/PERK pathway. Interestingly, bortezomib-resistant cell lines were noted to be more sensitive to the inhibition with CCF642-34 compared to bortezomib-naïve cells, which further supported the hypothesis of increased dependence of bortezomib-resistant MM cells on PDIA1. Further experiments showed synergism of CCF642-34 with bortezomib [286]. The CCF642-34 was then evaluated on a mouse xenograft with 50% of mice surviving beyond the end of the experiment (180 days). These extensive preclinical experiments highlighted the potential of PDIA1 inhibitors and further supported the strategy of targeting the PDIA1 isoform for the treatment of RRMM.

7.2. Peptidylprolyl Isomerase A

A recent single-arm phase 2 study (NCT04065789), assessed the combination of carfilzomib, daratumumab, lenalidomide, and dexamethasone in MM patients who failed to respond or experienced early relapse after a bortezomib-based induction regimen [289]. Single-cell RNA sequencing was applied to the participants’ samples, in order to explore the molecular pathways of MM resistance. The 41 MM patients of the trial were compared to 11 healthy individuals and 15 NDMM patients. Results showed upregulation of genes associated with endoplasmic reticulum stress pathways, such as peptidylprolyl isomerase A (PPIA), in patients with relapsed/refractory disease [289]. Cyclophilin A or PPIA is a critical enzyme in the protein folding response pathway, upregulation of which may represent an escape mechanism, and, therefore, the authors concluded that it can be a potential new therapeutic target for future drug development. Deletion of the PPIA gene or inhibition of PPIA with the use of a small molecule inhibitor, called ciclosporin, significantly sensitized the MM cells to PI.

7.3. Sec61 Translocon

Translocon is a protein of the membrane of the ER which mediates membrane insertion of most membrane proteins of organelles and facilitates translocation of almost all newly synthesized polypeptides destined for organelles, as well as most precursors of secretory proteins [290]. Sec61 is polypeptide-conducting channel, which is part of the translocon along with other proteins. Targeting the Sec61 of the translocon, thus disrupting transport of newly secreted or transmembrane proteins into the ER, leads to their degradation by the proteasome, which can eventually lead to cell apoptosis due to continuous proteotoxic stress response. The use of mycolactone, a small molecule that inhibits Sec61, along with PI was found to be effective in inducing apoptosis and synergistic in pre-clinical models of laboratory-derived MM cell lines [291]. This combination was found to cause enhanced ER stress with activation of pro-apoptotic UPR in MM cells, as reflected on transcript as well as protein levels. The synergy was maintained in bortezomib-resistant MM.1S cell lines, as well as in patient-derived MM cells. This target should be further explored as a potential novel therapeutic approach and drug development [291].

7.4. Cyclin-Dependent Kinase 6 (CKD6)

A recently published study performed integrated global quantitative tandem-mass-tag (TMT)-based proteomic and phosphoproteomic analyses in MM samples to identify a non-genetic resistance mechanism for IMiD that can be targeted by pharmacologic intervention. The results showed upregulation of CDK6, which appeared to decrease sensitivity to IMID [292]. Inhibition of CDK6 with palbociclib or degradation of CDK6 by PROTAC was found to be highly effective and synergistic with IMiD in pre-clinical in vivo and in vitro models [292]. These findings suggested further investigation of CDK6 as a potential drug target.

8. Conclusions

Despite the progress made to advance the therapeutic modalities in MM, treatment resistance and relapse continue to be a major issue. Hence, novel targeted and safer treatments are needed to combat this incurable disease. Understanding the genetic and epigenetic basis of the molecular evolution of MGUS to MM is key. For this purpose, using the power of next-generation sequencing coupled with genome editing will not only enable better prognostication, risk stratification, and prediction of therapy response, but, most importantly, will lead to the discovery of new strategies for prevention as well as curing of MM.

Author Contributions

Conceptualization, D.D. and B.K.J.; software, D.J.S; writing—original draft preparation, D.D.; writing—review and editing, D.J.S., F.U., D.J., M.H., H.S.S., J.K., J.P.M. and B.K.J.; supervision, B.K.J.; project administration, B.K.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was in part supported by the VeloSano Bike to Cure pilot award.

Acknowledgments

The BioRender website that was used for preparation of the figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Palumbo, A.; Anderson, K. Multiple myeloma. N. Engl. J. Med. 2011, 364, 1046–1060. [Google Scholar] [CrossRef] [PubMed]
  2. Short, K.D.; Rajkumar, S.V.; Larson, D.; Buadi, F.; Hayman, S.; Dispenzieri, A.; Gertz, M.; Kumar, S.; Mikhael, J.; Roy, V.; et al. Incidence of extramedullary disease in patients with multiple myeloma in the era of novel therapy, and the activity of pomalidomide on extramedullary myeloma. Leukemia 2011, 25, 906–908. [Google Scholar] [CrossRef] [PubMed]
  3. Varettoni, M.; Corso, A.; Pica, G.; Mangiacavalli, S.; Pascutto, C.; Lazzarino, M. Incidence, presenting features and outcome of extramedullary disease in multiple myeloma: A longitudinal study on 1003 consecutive patients. Ann. Oncol. 2010, 21, 325–330. [Google Scholar] [CrossRef] [PubMed]
  4. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef]
  5. Rajkumar, S.V.; Kumar, S. Multiple myeloma current treatment algorithms. Blood Cancer J. 2020, 10, 94. [Google Scholar] [CrossRef]
  6. Pinto, V.; Bergantim, R.; Caires, H.R.; Seca, H.; Guimarães, J.E.; Vasconcelos, M.H. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers 2020, 12, 407. [Google Scholar] [CrossRef]
  7. Dutta, A.K.; Fink, J.L.; Grady, J.P.; Morgan, G.J.; Mullighan, C.G.; To, L.B.; Hewett, D.R.; Zannettino, A.C.W. Subclonal evolution in disease progression from MGUS/SMM to multiple myeloma is characterised by clonal stability. Leukemia 2019, 33, 457–468. [Google Scholar] [CrossRef]
  8. Landgren, O.; Kyle, R.A.; Pfeiffer, R.M.; Katzmann, J.A.; Caporaso, N.E.; Hayes, R.B.; Dispenzieri, A.; Kumar, S.; Clark, R.J.; Baris, D.; et al. Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: A prospective study. Blood 2009, 113, 5412–5417. [Google Scholar] [CrossRef]
  9. Walker, B.A.; Mavrommatis, K.; Wardell, C.P.; Ashby, T.C.; Bauer, M.; Davies, F.E.; Rosenthal, A.; Wang, H.; Qu, P.; Hoering, A.; et al. Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood 2018, 132, 587–597. [Google Scholar] [CrossRef]
  10. Walker, B.A.; Boyle, E.M.; Wardell, C.P.; Murison, A.; Begum, D.B.; Dahir, N.M.; Proszek, P.Z.; Johnson, D.C.; Kaiser, M.F.; Melchor, L.; et al. Mutational Spectrum, Copy Number Changes, and Outcome: Results of a Sequencing Study of Patients With Newly Diagnosed Myeloma. J. Clin. Oncol. 2015, 33, 3911–3920. [Google Scholar] [CrossRef]
  11. Melchor, L.; Brioli, A.; Wardell, C.P.; Murison, A.; Potter, N.E.; Kaiser, M.F.; Fryer, R.A.; Johnson, D.C.; Begum, D.B.; Hulkki Wilson, S.; et al. Single-cell genetic analysis reveals the composition of initiating clones and phylogenetic patterns of branching and parallel evolution in myeloma. Leukemia 2014, 28, 1705–1715. [Google Scholar] [CrossRef] [PubMed]
  12. Maura, F.; Bolli, N.; Angelopoulos, N.; Dawson, K.J.; Leongamornlert, D.; Martincorena, I.; Mitchell, T.J.; Fullam, A.; Gonzalez, S.; Szalat, R.; et al. Genomic landscape and chronological reconstruction of driver events in multiple myeloma. Nat. Commun. 2019, 10, 3835. [Google Scholar] [CrossRef] [PubMed]
  13. Rasche, L.; Chavan, S.S.; Stephens, O.W.; Patel, P.H.; Tytarenko, R.; Ashby, C.; Bauer, M.; Stein, C.; Deshpande, S.; Wardell, C.; et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat. Commun. 2017, 8, 268. [Google Scholar] [CrossRef] [PubMed]
  14. Bahlis, N.J. Darwinian evolution and tiding clones in multiple myeloma. Blood 2012, 120, 927–928. [Google Scholar] [CrossRef]
  15. Maura, F.; Bolli, N.; Rustad, E.H.; Hultcrantz, M.; Munshi, N.; Landgren, O. Moving From Cancer Burden to Cancer Genomics for Smoldering Myeloma: A Review. JAMA Oncol. 2020, 6, 425–432. [Google Scholar] [CrossRef]
  16. Kyle, R.A.; Larson, D.R.; Therneau, T.M.; Dispenzieri, A.; Kumar, S.; Cerhan, J.R.; Rajkumar, S.V. Long-Term Follow-up of Monoclonal Gammopathy of Undetermined Significance. N. Engl. J. Med. 2018, 378, 241–249. [Google Scholar] [CrossRef]
  17. Landgren, O.; Hofmann, J.N.; McShane, C.M.; Santo, L.; Hultcrantz, M.; Korde, N.; Mailankody, S.; Kazandjian, D.; Murata, K.; Thoren, K.; et al. Association of Immune Marker Changes with Progression of Monoclonal Gammopathy of Undetermined Significance to Multiple Myeloma. JAMA Oncol. 2019, 5, 1293–1301. [Google Scholar] [CrossRef]
  18. Mailankody, S.; Kazandjian, D.; Korde, N.; Roschewski, M.; Manasanch, E.; Bhutani, M.; Tageja, N.; Kwok, M.; Zhang, Y.; Zingone, A.; et al. Baseline mutational patterns and sustained MRD negativity in patients with high-risk smoldering myeloma. Blood Adv. 2017, 1, 1911–1918. [Google Scholar] [CrossRef]
  19. Chapman, M.A.; Lawrence, M.S.; Keats, J.J.; Cibulskis, K.; Sougnez, C.; Schinzel, A.C.; Harview, C.L.; Brunet, J.P.; Ahmann, G.J.; Adli, M.; et al. Initial genome sequencing and analysis of multiple myeloma. Nature 2011, 471, 467–472. [Google Scholar] [CrossRef]
  20. Lohr, J.G.; Stojanov, P.; Carter, S.L.; Cruz-Gordillo, P.; Lawrence, M.S.; Auclair, D.; Sougnez, C.; Knoechel, B.; Gould, J.; Saksena, G.; et al. Widespread genetic heterogeneity in multiple myeloma: Implications for targeted therapy. Cancer Cell 2014, 25, 91–101. [Google Scholar] [CrossRef]
  21. Bolli, N.; Avet-Loiseau, H.; Wedge, D.C.; Van Loo, P.; Alexandrov, L.B.; Martincorena, I.; Dawson, K.J.; Iorio, F.; Nik-Zainal, S.; Bignell, G.R.; et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat. Commun. 2014, 5, 2997. [Google Scholar] [CrossRef] [PubMed]
  22. Greipp, P.R.; San Miguel, J.; Durie, B.G.; Crowley, J.J.; Barlogie, B.; Bladé, J.; Boccadoro, M.; Child, J.A.; Avet-Loiseau, H.; Kyle, R.A.; et al. International staging system for multiple myeloma. J. Clin. Oncol. 2005, 23, 3412–3420. [Google Scholar] [CrossRef] [PubMed]
  23. Palumbo, A.; Avet-Loiseau, H.; Oliva, S.; Lokhorst, H.M.; Goldschmidt, H.; Rosinol, L.; Richardson, P.; Caltagirone, S.; Lahuerta, J.J.; Facon, T.; et al. Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group. J. Clin. Oncol. 2015, 33, 2863–2869. [Google Scholar] [CrossRef] [PubMed]
  24. Walker, B.A.; Mavrommatis, K.; Wardell, C.P.; Ashby, T.C.; Bauer, M.; Davies, F.; Rosenthal, A.; Wang, H.; Qu, P.; Hoering, A.; et al. A high-risk, Double-Hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia 2019, 33, 159–170. [Google Scholar] [CrossRef] [PubMed]
  25. Van Laar, R.; Flinchum, R.; Brown, N.; Ramsey, J.; Riccitelli, S.; Heuck, C.; Barlogie, B.; Shaughnessy, J.D., Jr. Translating a gene expression signature for multiple myeloma prognosis into a robust high-throughput assay for clinical use. BMC Med. Genom. 2014, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  26. Kuiper, R.; Broyl, A.; de Knegt, Y.; van Vliet, M.H.; van Beers, E.H.; van der Holt, B.; el Jarari, L.; Mulligan, G.; Gregory, W.; Morgan, G.; et al. A gene expression signature for high-risk multiple myeloma. Leukemia 2012, 26, 2406–2413. [Google Scholar] [CrossRef]
  27. Shah, V.; Sherborne, A.L.; Johnson, D.C.; Ellis, S.; Price, A.; Chowdhury, F.; Kendall, J.; Jenner, M.W.; Drayson, M.T.; Owen, R.G.; et al. Predicting ultrahigh risk multiple myeloma by molecular profiling: An analysis of newly diagnosed transplant eligible myeloma XI trial patients. Leukemia 2020, 34, 3091–3096. [Google Scholar] [CrossRef]
  28. Poczta, A.; Rogalska, A.; Marczak, A. Treatment of Multiple Myeloma and the Role of Melphalan in the Era of Modern Therapies-Current Research and Clinical Approaches. J. Clin. Med. 2021, 10, 1841. [Google Scholar] [CrossRef]
  29. Schjesvold, F.; Oriol, A. Current and Novel Alkylators in Multiple Myeloma. Cancers 2021, 13, 2465. [Google Scholar] [CrossRef]
  30. Mateos, M.V.; Bladé, J.; Bringhen, S.; Ocio, E.M.; Efebera, Y.; Pour, L.; Gay, F.; Sonneveld, P.; Gullbo, J.; Richardson, P.G. Melflufen: A Peptide-Drug Conjugate for the Treatment of Multiple Myeloma. J. Clin. Med. 2020, 9, 3120. [Google Scholar] [CrossRef]
  31. 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] [PubMed]
  32. Lu, G.; Middleton, R.E.; Sun, H.; Naniong, M.; Ott, C.J.; Mitsiades, C.S.; Wong, K.K.; Bradner, J.E.; Kaelin, W.G., Jr. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 2014, 343, 305–309. [Google Scholar] [CrossRef] [PubMed]
  33. Krönke, J.; Udeshi, N.D.; Narla, A.; Grauman, P.; Hurst, S.N.; McConkey, M.; Svinkina, T.; Heckl, D.; Comer, E.; Li, X.; et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 2014, 343, 301–305. [Google Scholar] [CrossRef] [PubMed]
  34. 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]
  35. Hayashi, T.; Hideshima, T.; Akiyama, M.; Podar, K.; Yasui, H.; Raje, N.; Kumar, S.; Chauhan, D.; Treon, S.P.; Richardson, P.; et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: Clinical application. Br. J. Haematol. 2005, 128, 192–203. [Google Scholar] [CrossRef] [PubMed]
  36. Fedele, P.L.; Willis, S.N.; Liao, Y.; Low, M.S.; Rautela, J.; Segal, D.H.; Gong, J.N.; Huntington, N.D.; Shi, W.; Huang, D.C.S.; et al. IMiDs prime myeloma cells for daratumumab-mediated cytotoxicity through loss of Ikaros and Aiolos. Blood 2018, 132, 2166–2178. [Google Scholar] [CrossRef]
  37. Hsu, A.K.; Quach, H.; Tai, T.; Prince, H.M.; Harrison, S.J.; Trapani, J.A.; Smyth, M.J.; Neeson, P.; Ritchie, D.S. The immunostimulatory effect of lenalidomide on NK-cell function is profoundly inhibited by concurrent dexamethasone therapy. Blood 2011, 117, 1605–1613. [Google Scholar] [CrossRef]
  38. Hideshima, T.; Ogiya, D.; Liu, J.; Harada, T.; Kurata, K.; Bae, J.; Massefski, W.; Anderson, K.C. Immunomodulatory drugs activate NK cells via both Zap-70 and cereblon-dependent pathways. Leukemia 2021, 35, 177–188. [Google Scholar] [CrossRef]
  39. 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]
  40. Reddy, N.; Hernandez-Ilizaliturri, F.J.; Deeb, G.; Roth, M.; Vaughn, M.; Knight, J.; Wallace, P.; Czuczman, M.S. Immunomodulatory drugs stimulate natural killer-cell function, alter cytokine production by dendritic cells, and inhibit angiogenesis enhancing the anti-tumour activity of rituximab in vivo. Br. J. Haematol. 2008, 140, 36–45. [Google Scholar] [CrossRef]
  41. Thakurta, A.; Pierceall, W.E.; Amatangelo, M.D.; Flynt, E.; Agarwal, A. Developing next generation immunomodulatory drugs and their combinations in multiple myeloma. Oncotarget 2021, 12, 1555–1563. [Google Scholar] [CrossRef] [PubMed]
  42. Bjorklund, C.C.; Kang, J.; Amatangelo, M.; Polonskaia, A.; Katz, M.; Chiu, H.; Couto, S.; Wang, M.; Ren, Y.; Ortiz, M.; et al. Iberdomide (CC-220) is a potent cereblon E3 ligase modulator with antitumor and immunostimulatory activities in lenalidomide- and pomalidomide-resistant multiple myeloma cells with dysregulated CRBN. Leukemia 2020, 34, 1197–1201. [Google Scholar] [CrossRef] [PubMed]
  43. Lonial, S.; Donk, N.W.C.J.v.d.; Popat, R.; Zonder, J.A.; Minnema, M.C.; Larsen, J.; Nguyen, T.V.; Chen, M.S.; Bensmaine, A.; Cota, M.; et al. First clinical (phase 1b/2a) study of iberdomide (CC-220; IBER), a CELMoD, in combination with dexamethasone (DEX) in patients (pts) with relapsed/refractory multiple myeloma (RRMM). J. Clin. Oncol. 2019, 37, 8006. [Google Scholar] [CrossRef]
  44. Van de Donk, N.W.C.J.; Popat, R.; Larsen, J.; Minnema, M.C.; Jagannath, S.; Oriol, A.; Zonder, J.; Richardson, P.G.; Rodriguez-Otero, P.; Badros, A.Z.; et al. First Results of Iberdomide (IBER.; CC-220) in Combination with Dexamethasone (DEX) and Daratumumab (DARA) or Bortezomib (BORT) in Patients with Relapsed/Refractory Multiple Myeloma (RRMM). Blood 2020, 136, 16–17. [Google Scholar] [CrossRef]
  45. Lopez-Girona, A.; Havens, C.G.; Lu, G.; Rychak, E.; Mendy, D.; Gaffney, B.; Surka, C.; Lu, C.-C.; Matyskiela, M.; Khambatta, G.; et al. CC-92480 Is a Novel Cereblon E3 Ligase Modulator with Enhanced Tumoricidal and Immunomodulatory Activity Against Sensitive and Resistant Multiple Myeloma Cells. Blood 2019, 134, 1812. [Google Scholar] [CrossRef]
  46. Richardson, P.G.; Vangsted, A.J.; Ramasamy, K.; Trudel, S.; Martínez, J.; Mateos, M.-V.; Otero, P.R.; Lonial, S.; Popat, R.; Oriol, A.; et al. First-in-human phase I study of the novel CELMoD agent CC-92480 combined with dexamethasone (DEX) in patients (pts) with relapsed/refractory multiple myeloma (RRMM). J. Clin. Oncol. 2020, 38, 8500. [Google Scholar] [CrossRef]
  47. Rasco, D.W.; Papadopoulos, K.P.; Pourdehnad, M.; Gandhi, A.K.; Hagner, P.R.; Li, Y.; Wei, X.; Chopra, R.; Hege, K.; DiMartino, J.; et al. A First-in-Human Study of Novel Cereblon Modulator Avadomide (CC-122) in Advanced Malignancies. Clin. Cancer Res. 2019, 25, 90–98. [Google Scholar] [CrossRef]
  48. Berdeja, J.; Ailawadhi, S.; Horwitz, S.M.; Matous, J.V.; Mehta-Shah, N.; Martin, T.; Muchtar, E.; Richardson, P.G.; Richard, S.; Bhutani, M.; et al. A Phase 1 Study of CFT7455, a Novel Degrader of IKZF1/3, in Multiple Myeloma and Non-Hodgkin Lymphoma. Blood 2021, 138, 1675. [Google Scholar] [CrossRef]
  49. McConkey, D.J.; Zhu, K. Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist. Updat. 2008, 11, 164–179. [Google Scholar] [CrossRef]
  50. Hideshima, T.; Richardson, P.G.; Anderson, K.C. Mechanism of action of proteasome inhibitors and deacetylase inhibitors and the biological basis of synergy in multiple myeloma. Mol. Cancer Ther. 2011, 10, 2034–2042. [Google Scholar] [CrossRef]
  51. Gandolfi, S.; Laubach, J.P.; Hideshima, T.; Chauhan, D.; Anderson, K.C.; Richardson, P.G. The proteasome and proteasome inhibitors in multiple myeloma. Cancer Metastasis Rev. 2017, 36, 561–584. [Google Scholar] [CrossRef] [PubMed]
  52. Gulla, A.; Morelli, E.; Samur, M.K.; Botta, C.; Hideshima, T.; Bianchi, G.; Fulciniti, M.; Malvestiti, S.; Prabhala, R.H.; Talluri, S.; et al. Bortezomib induces anti-multiple myeloma immune response mediated by cGAS/STING pathway activation. Blood Cancer Discov. 2021, 2, 468–483. [Google Scholar] [CrossRef] [PubMed]
  53. Spisek, R.; Charalambous, A.; Mazumder, A.; Vesole, D.H.; Jagannath, S.; Dhodapkar, M.V. Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: Therapeutic implications. Blood 2007, 109, 4839–4845. [Google Scholar] [CrossRef]
  54. Shi, J.; Tricot, G.J.; Garg, T.K.; Malaviarachchi, P.A.; Szmania, S.M.; Kellum, R.E.; Storrie, B.; Mulder, A.; Shaughnessy, J.D., Jr.; Barlogie, B.; et al. Bortezomib down-regulates the cell-surface expression of HLA class I and enhances natural killer cell-mediated lysis of myeloma. Blood 2008, 111, 1309–1317. [Google Scholar] [CrossRef]
  55. Ito, S. Proteasome Inhibitors for the Treatment of Multiple Myeloma. Cancers 2020, 12, 265. [Google Scholar] [CrossRef]
  56. Fostier, K.; De Becker, A.; Schots, R. Carfilzomib: A novel treatment in relapsed and refractory multiple myeloma. OncoTargets Ther. 2012, 5, 237–244. [Google Scholar] [CrossRef]
  57. Gupta, N.; Hanley, M.J.; Xia, C.; Labotka, R.; Harvey, R.D.; Venkatakrishnan, K. Clinical Pharmacology of Ixazomib: The First Oral Proteasome Inhibitor. Clin. Pharmacokinet. 2019, 58, 431–449. [Google Scholar] [CrossRef]
  58. Kubiczkova, L.; Pour, L.; Sedlarikova, L.; Hajek, R.; Sevcikova, S. Proteasome inhibitors—Molecular basis and current perspectives in multiple myeloma. J. Cell Mol. Med. 2014, 18, 947–961. [Google Scholar] [CrossRef]
  59. Potts, B.C.; Albitar, M.X.; Anderson, K.C.; Baritaki, S.; Berkers, C.; Bonavida, B.; Chandra, J.; Chauhan, D.; Cusack, J.C., Jr.; Fenical, W.; et al. Marizomib, a proteasome inhibitor for all seasons: Preclinical profile and a framework for clinical trials. Curr. Cancer Drug Targets 2011, 11, 254–284. [Google Scholar] [CrossRef]
  60. Hari, P.; Matous, J.V.; Voorhees, P.M.; Shain, K.H.; Obreja, M.; Frye, J.; Fujii, H.; Jakubowiak, A.J.; Rossi, D.; Sonneveld, P. Oprozomib in patients with newly diagnosed multiple myeloma. Blood Cancer J. 2019, 9, 66. [Google Scholar] [CrossRef]
  61. Hari, P.; Paba-Prada, C.E.; Voorhees, P.M.; Frye, J.; Chang, Y.L.; Moreau, P.; Zonder, J.; Boccia, R.; Shain, K.H. Efficacy and safety results from a phase 1b/2, multicenter, open-label study of oprozomib and dexamethasone in patients with relapsed and/or refractory multiple myeloma. Leuk Res. 2019, 83, 106172. [Google Scholar] [CrossRef] [PubMed]
  62. Park, J.; Park, E.; Jung, C.K.; Kang, S.W.; Kim, B.G.; Jung, Y.; Kim, T.H.; Lim, J.Y.; Lee, S.E.; Min, C.K.; et al. Oral proteasome inhibitor with strong preclinical efficacy in myeloma models. BMC Cancer 2016, 16, 247. [Google Scholar] [CrossRef] [PubMed]
  63. Swamydas, M.; Murphy, E.V.; Ignatz-Hoover, J.J.; Malek, E.; Driscoll, J.J. Deciphering mechanisms of immune escape to inform immunotherapeutic strategies in multiple myeloma. J. Hematol. Oncol. 2022, 15, 17. [Google Scholar] [CrossRef] [PubMed]
  64. Franssen, L.E.; Mutis, T.; Lokhorst, H.M.; van de Donk, N. Immunotherapy in myeloma: How far have we come? Ther. Adv. Hematol. 2019, 10, 2040620718822660. [Google Scholar] [CrossRef]
  65. Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef]
  66. Vyas, M.; Müller, R.; Pogge von Strandmann, E. Antigen Loss Variants: Catching Hold of Escaping Foes. Front. Immunol. 2017, 8, 175. [Google Scholar] [CrossRef]
  67. Lozano, E.; Díaz, T.; Mena, M.P.; Suñe, G.; Calvo, X.; Calderón, M.; Pérez-Amill, L.; Rodríguez, V.; Pérez-Galán, P.; Roué, G.; et al. Loss of the Immune Checkpoint CD85j/LILRB1 on Malignant Plasma Cells Contributes to Immune Escape in Multiple Myeloma. J. Immunol. 2018, 200, 2581–2591. [Google Scholar] [CrossRef]
  68. Dhatchinamoorthy, K.; Colbert, J.D.; Rock, K.L. Cancer Immune Evasion Through Loss of MHC Class I Antigen Presentation. Front. Immunol. 2021, 12, 636568. [Google Scholar] [CrossRef]
  69. Gulla, A.; Morelli, E.; Samur, M.K.; Botta, C.; Johnstone, M.; Bianchi, G.; Fulciniti, M.; Yamamoto, L.; Prabhala, R.; Wen, K.; et al. Gabarap Loss Mediates Immune Escape in High Risk Multiple Myeloma. Blood 2021, 138, 891. [Google Scholar] [CrossRef]
  70. Racanelli, V.; Leone, P.; Frassanito, M.A.; Brunetti, C.; Perosa, F.; Ferrone, S.; Dammacco, F. Alterations in the antigen processing-presenting machinery of transformed plasma cells are associated with reduced recognition by CD8+ T cells and characterize the progression of MGUS to multiple myeloma. Blood 2010, 115, 1185–1193. [Google Scholar] [CrossRef]
  71. Kumar, S.; Kimlinger, T.; Morice, W. Immunophenotyping in multiple myeloma and related plasma cell disorders. Best Pract. Res. Clin. Haematol. 2010, 23, 433–451. [Google Scholar] [CrossRef] [PubMed]
  72. Dwivedi, S.; Rendón-Huerta, E.P.; Ortiz-Navarrete, V.; Montaño, L.F. CD38 and Regulation of the Immune Response Cells in Cancer. J. Oncol. 2021, 2021, 6630295. [Google Scholar] [CrossRef] [PubMed]
  73. Maecker, B.; Anderson, K.S.; von Bergwelt-Baildon, M.S.; Weller, E.; Vonderheide, R.H.; Richardson, P.G.; Schlossman, R.L.; Menezes, I.A.; Xia, Z.; Munshi, N.C.; et al. Viral antigen-specific CD8+ T-cell responses are impaired in multiple myeloma. Br. J. Haematol. 2003, 121, 842–848. [Google Scholar] [CrossRef] [PubMed]
  74. Dhodapkar, M.V.; Krasovsky, J.; Osman, K.; Geller, M.D. Vigorous premalignancy-specific effector T cell response in the bone marrow of patients with monoclonal gammopathy. J. Exp. Med. 2003, 198, 1753–1757. [Google Scholar] [CrossRef] [PubMed]
  75. Suen, H.; Brown, R.; Yang, S.; Weatherburn, C.; Ho, P.J.; Woodland, N.; Nassif, N.; Barbaro, P.; Bryant, C.; Hart, D.; et al. Multiple myeloma causes clonal T-cell immunosenescence: Identification of potential novel targets for promoting tumour immunity and implications for checkpoint blockade. Leukemia 2016, 30, 1716–1724. [Google Scholar] [CrossRef]
  76. 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]
  77. Bernal, M.; Garrido, P.; Jiménez, P.; Carretero, R.; Almagro, M.; López, P.; Navarro, P.; Garrido, F.; Ruiz-Cabello, F. Changes in activatory and inhibitory natural killer (NK) receptors may induce progression to multiple myeloma: Implications for tumor evasion of T and NK cells. Hum. Immunol. 2009, 70, 854–857. [Google Scholar] [CrossRef]
  78. Jinushi, M.; Vanneman, M.; Munshi, N.C.; Tai, Y.T.; Prabhala, R.H.; Ritz, J.; Neuberg, D.; Anderson, K.C.; Carrasco, D.R.; Dranoff, G. MHC class I chain-related protein A antibodies and shedding are associated with the progression of multiple myeloma. Proc. Natl. Acad. Sci. USA 2008, 105, 1285–1290. [Google Scholar] [CrossRef]
  79. De Jong, M.M.E.; Kellermayer, Z.; Papazian, N.; Tahri, S.; Hofste Op Bruinink, D.; Hoogenboezem, R.; Sanders, M.A.; van de Woestijne, P.C.; Bos, P.K.; Khandanpour, C.; et al. The multiple myeloma microenvironment is defined by an inflammatory stromal cell landscape. Nat. Immunol. 2021, 22, 769–780. [Google Scholar] [CrossRef]
  80. Prabhala, R.H.; Neri, P.; Bae, J.E.; Tassone, P.; Shammas, M.A.; Allam, C.K.; Daley, J.F.; Chauhan, D.; Blanchard, E.; Thatte, H.S.; et al. Dysfunctional T regulatory cells in multiple myeloma. Blood 2006, 107, 301–304. [Google Scholar] [CrossRef]
  81. 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] [PubMed]
  82. Banerjee, D.K.; Dhodapkar, M.V.; Matayeva, E.; Steinman, R.M.; Dhodapkar, K.M. Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients. Blood 2006, 108, 2655–2661. [Google Scholar] [CrossRef] [PubMed]
  83. 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] [PubMed]
  84. Dima, D.; Dower, J.; Comenzo, R.L.; Varga, C. Evaluating Daratumumab in the Treatment of Multiple Myeloma: Safety, Efficacy and Place in Therapy. Cancer Manag. Res. 2020, 12, 7891–7903. [Google Scholar] [CrossRef] [PubMed]
  85. Lee, H.C. Structure and enzymatic functions of human CD38. Mol. Med. 2006, 12, 317–323. [Google Scholar] [CrossRef]
  86. Horenstein, A.L.; Quarona, V.; Toscani, D.; Costa, F.; Chillemi, A.; Pistoia, V.; Giuliani, N.; Malavasi, F. Adenosine Generated in the Bone Marrow Niche Through a CD38-Mediated Pathway Correlates with Progression of Human Myeloma. Mol. Med. 2016, 22, 694–704. [Google Scholar] [CrossRef]
  87. Horenstein, A.L.; Chillemi, A.; Zaccarello, G.; Bruzzone, S.; Quarona, V.; Zito, A.; Serra, S.; Malavasi, F. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2013, 2, e26246. [Google Scholar] [CrossRef]
  88. Hogan, K.A.; Chini, C.C.S.; Chini, E.N. The Multi-faceted Ecto-enzyme CD38: Roles in Immunomodulation, Cancer, Aging, and Metabolic Diseases. Front. Immunol. 2019, 10, 1187. [Google Scholar] [CrossRef]
  89. De Weers, M.; Tai, Y.T.; van der Veer, M.S.; Bakker, J.M.; Vink, T.; Jacobs, D.C.; Oomen, L.A.; Peipp, M.; Valerius, T.; Slootstra, J.W.; et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J. Immunol. 2011, 186, 1840–1848. [Google Scholar] [CrossRef]
  90. Overdijk, M.B.; Jansen, J.H.; Nederend, M.; Lammerts van Bueren, J.J.; Groen, R.W.; Parren, P.W.; Leusen, J.H.; Boross, P. The Therapeutic CD38 Monoclonal Antibody Daratumumab Induces Programmed Cell Death via Fcγ Receptor-Mediated Cross-Linking. J. Immunol. 2016, 197, 807–813. [Google Scholar] [CrossRef]
  91. Overdijk, M.B.; Verploegen, S.; Bögels, M.; van Egmond, M.; Lammerts van Bueren, J.J.; Mutis, T.; Groen, R.W.; Breij, E.; Martens, A.C.; Bleeker, W.K.; et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. MAbs 2015, 7, 311–321. [Google Scholar] [CrossRef] [PubMed]
  92. Krejcik, J.; Casneuf, T.; Nijhof, I.S.; Verbist, B.; Bald, J.; Plesner, T.; Syed, K.; Liu, K.; van de Donk, N.W.; Weiss, B.M.; et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood 2016, 128, 384–394. [Google Scholar] [CrossRef] [PubMed]
  93. Lokhorst, H.M.; Plesner, T.; Laubach, J.P.; Nahi, H.; Gimsing, P.; Hansson, M.; Minnema, M.C.; Lassen, U.; Krejcik, J.; Palumbo, A.; et al. Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. N. Engl. J. Med. 2015, 373, 1207–1219. [Google Scholar] [CrossRef] [PubMed]
  94. Lonial, S.; Weiss, B.M.; Usmani, S.Z.; Singhal, S.; Chari, A.; Bahlis, N.J.; Belch, A.; Krishnan, A.; Vescio, R.A.; Mateos, M.V.; et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): An open-label, randomised, phase 2 trial. Lancet 2016, 387, 1551–1560. [Google Scholar] [CrossRef]
  95. Usmani, S.Z.; Weiss, B.M.; Plesner, T.; Bahlis, N.J.; Belch, A.; Lonial, S.; Lokhorst, H.M.; Voorhees, P.M.; Richardson, P.G.; Chari, A.; et al. Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. Blood 2016, 128, 37–44. [Google Scholar] [CrossRef]
  96. Palumbo, A.; Chanan-Khan, A.; Weisel, K.; Nooka, A.K.; Masszi, T.; Beksac, M.; Spicka, I.; Hungria, V.; Munder, M.; Mateos, M.V.; et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2016, 375, 754–766. [Google Scholar] [CrossRef]
  97. Van der Veer, M.S.; de Weers, M.; van Kessel, B.; Bakker, J.M.; Wittebol, S.; Parren, P.W.; Lokhorst, H.M.; Mutis, T. Towards effective immunotherapy of myeloma: Enhanced elimination of myeloma cells by combination of lenalidomide with the human CD38 monoclonal antibody daratumumab. Haematologica 2011, 96, 284–290. [Google Scholar] [CrossRef]
  98. Deckert, J.; Wetzel, M.C.; Bartle, L.M.; Skaletskaya, A.; Goldmacher, V.S.; Vallée, F.; Zhou-Liu, Q.; Ferrari, P.; Pouzieux, S.; Lahoute, C.; et al. SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clin. Cancer Res. 2014, 20, 4574–4583. [Google Scholar] [CrossRef]
  99. Van de Donk, N.W.; Moreau, P.; Plesner, T.; Palumbo, A.; Gay, F.; Laubach, J.P.; Malavasi, F.; Avet-Loiseau, H.; Mateos, M.V.; Sonneveld, P.; et al. Clinical efficacy and management of monoclonal antibodies targeting CD38 and SLAMF7 in multiple myeloma. Blood 2016, 127, 681–695. [Google Scholar] [CrossRef]
  100. Attal, M.; Richardson, P.G.; Rajkumar, S.V.; San-Miguel, J.; Beksac, M.; Spicka, I.; Leleu, X.; Schjesvold, F.; Moreau, P.; Dimopoulos, M.A.; et al. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): A randomised, multicentre, open-label, phase 3 study. Lancet 2019, 394, 2096–2107. [Google Scholar] [CrossRef]
  101. Moreau, P.; Dimopoulos, M.A.; Mikhael, J.; Yong, K.; Capra, M.; Facon, T.; Hajek, R.; Špička, I.; Baker, R.; Kim, K.; et al. Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): A multicentre, open-label, randomised phase 3 trial. Lancet 2021, 397, 2361–2371. [Google Scholar] [CrossRef]
  102. Veillette, A.; Guo, H. CS1, a SLAM family receptor involved in immune regulation, is a therapeutic target in multiple myeloma. Crit. Rev. Oncol. Hematol. 2013, 88, 168–177. [Google Scholar] [CrossRef] [PubMed]
  103. Richardson, P.G.; Lonial, S.; Jakubowiak, A.J.; Harousseau, J.L.; Anderson, K.C. Monoclonal antibodies in the treatment of multiple myeloma. Br. J. Haematol. 2011, 154, 745–754. [Google Scholar] [CrossRef] [PubMed]
  104. Hofmeister, C.C.; Lonial, S. How to Integrate Elotuzumab and Daratumumab Into Therapy for Multiple Myeloma. J. Clin. Oncol. 2016, 34, 4421–4430. [Google Scholar] [CrossRef] [PubMed]
  105. Kumaresan, P.R.; Lai, W.C.; Chuang, S.S.; Bennett, M.; Mathew, P.A. CS1, a novel member of the CD2 family, is homophilic and regulates NK cell function. Mol. Immunol. 2002, 39, 1–8. [Google Scholar] [CrossRef]
  106. Tai, Y.T.; Dillon, M.; Song, W.; Leiba, M.; Li, X.F.; Burger, P.; Lee, A.I.; Podar, K.; Hideshima, T.; Rice, A.G.; et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood 2008, 112, 1329–1337. [Google Scholar] [CrossRef]
  107. Wang, Y.; Sanchez, L.; Siegel, D.S.; Wang, M.L. Elotuzumab for the treatment of multiple myeloma. J. Hematol. Oncol. 2016, 9, 55. [Google Scholar] [CrossRef]
  108. Pazina, T.; James, A.M.; MacFarlane, A.W.t.; Bezman, N.A.; Henning, K.A.; Bee, C.; Graziano, R.F.; Robbins, M.D.; Cohen, A.D.; Campbell, K.S. The anti-SLAMF7 antibody elotuzumab mediates NK cell activation through both CD16-dependent and -independent mechanisms. Oncoimmunology 2017, 6, e1339853. [Google Scholar] [CrossRef]
  109. Hsi, E.D.; Steinle, R.; Balasa, B.; Szmania, S.; Draksharapu, A.; Shum, B.P.; Huseni, M.; Powers, D.; Nanisetti, A.; Zhang, Y.; et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin. Cancer Res. 2008, 14, 2775–2784. [Google Scholar] [CrossRef]
  110. Sola, C.; Blery, M.; Bonnafous, C.; Bonnet, E.; Fuseri, N.; Graziano, R.F.; Morel, Y.; André, P. Lirilumab Enhances Anti-Tumor Efficacy of Elotuzumab. Blood 2014, 124, 4711. [Google Scholar] [CrossRef]
  111. Robbins, M.; Jure-Kunkel, M.; Dito, G.; Andre, P.; Zhang, H.-f.; Bezman, N.; Graziano, R.F. Effects of IL-21, KIR Blockade, and CD137 Agonism on the Non-Clinical Activity of Elotuzumab. Blood 2014, 124, 4717. [Google Scholar] [CrossRef]
  112. Collins, S.M.; Bakan, C.E.; Swartzel, G.D.; Hofmeister, C.C.; Efebera, Y.A.; Kwon, H.; Starling, G.C.; Ciarlariello, D.; Bhaskar, S.; Briercheck, E.L.; et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: Evidence for augmented NK cell function complementing ADCC. Cancer Immunol. Immunother. 2013, 62, 1841–1849. [Google Scholar] [CrossRef]
  113. Van Rhee, F.; Szmania, S.M.; Dillon, M.; van Abbema, A.M.; Li, X.; Stone, M.K.; Garg, T.K.; Shi, J.; Moreno-Bost, A.M.; Yun, R.; et al. Combinatorial efficacy of anti-CS1 monoclonal antibody elotuzumab (HuLuc63) and bortezomib against multiple myeloma. Mol. Cancer Ther. 2009, 8, 2616–2624. [Google Scholar] [CrossRef]
  114. Balasa, B.; Yun, R.; Belmar, N.A.; Fox, M.; Chao, D.T.; Robbins, M.D.; Starling, G.C.; Rice, A.G. Elotuzumab enhances natural killer cell activation and myeloma cell killing through interleukin-2 and TNF-α pathways. Cancer Immunol. Immunother. 2015, 64, 61–73. [Google Scholar] [CrossRef]
  115. Lonial, S.; Dimopoulos, M.; Palumbo, A.; White, D.; Grosicki, S.; Spicka, I.; Walter-Croneck, A.; Moreau, P.; Mateos, M.V.; Magen, H.; et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2015, 373, 621–631. [Google Scholar] [CrossRef]
  116. 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]
  117. Dimopoulos, M.A.; Lonial, S.; Betts, K.A.; Chen, C.; Zichlin, M.L.; Brun, A.; Signorovitch, J.E.; Makenbaeva, D.; Mekan, S.; Sy, O.; et al. Elotuzumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: Extended 4-year follow-up and analysis of relative progression-free survival from the randomized ELOQUENT-2 trial. Cancer 2018, 124, 4032–4043. [Google Scholar] [CrossRef]
  118. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef]
  119. Zou, W.; Chen, L. Inhibitory B7-family molecules in the tumour microenvironment. Nat. Rev. Immunol. 2008, 8, 467–477. [Google Scholar] [CrossRef]
  120. Kwon, M.; Kim, C.G.; Lee, H.; Cho, H.; Kim, Y.; Lee, E.C.; Choi, S.J.; Park, J.; Seo, I.H.; Bogen, B.; et al. PD-1 Blockade Reinvigorates Bone Marrow CD8(+) T Cells from Patients with Multiple Myeloma in the Presence of TGFβ Inhibitors. Clin. Cancer Res. 2020, 26, 1644–1655. [Google Scholar] [CrossRef]
  121. Benson, D.M., Jr.; Bakan, C.E.; Mishra, A.; Hofmeister, C.C.; Efebera, Y.; Becknell, B.; Baiocchi, R.A.; Zhang, J.; Yu, J.; Smith, M.K.; et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: A therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 2010, 116, 2286–2294. [Google Scholar] [CrossRef]
  122. Liu, J.; Hamrouni, A.; Wolowiec, D.; Coiteux, V.; Kuliczkowski, K.; Hetuin, D.; Saudemont, A.; Quesnel, B. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007, 110, 296–304. [Google Scholar] [CrossRef]
  123. Tamura, H.; Ishibashi, M.; Yamashita, T.; Tanosaki, S.; Okuyama, N.; Kondo, A.; Hyodo, H.; Shinya, E.; Takahashi, H.; Dong, H.; et al. Marrow stromal cells induce B7-H1 expression on myeloma cells, generating aggressive characteristics in multiple myeloma. Leukemia 2013, 27, 464–472. [Google Scholar] [CrossRef]
  124. Mussetti, A.; Pellegrinelli, A.; Cieri, N.; Garzone, G.; Dominoni, F.; Cabras, A.; Montefusco, V. PD-L1, LAG3, and HLA-DR are increasingly expressed during smoldering myeloma progression. Ann. Hematol. 2019, 98, 1713–1720. [Google Scholar] [CrossRef]
  125. Lucas, F.; Pennell, M.; Huang, Y.; Benson, D.M.; Efebera, Y.A.; Chaudhry, M.; Hughes, T.; Woyach, J.A.; Byrd, J.C.; Zhang, S.; et al. T Cell Transcriptional Profiling and Immunophenotyping Uncover LAG3 as a Potential Significant Target of Immune Modulation in Multiple Myeloma. Biol. Blood Marrow Transplant. 2020, 26, 7–15. [Google Scholar] [CrossRef]
  126. Guillerey, C.; Harjunpää, H.; Carrié, N.; Kassem, S.; Teo, T.; Miles, K.; Krumeich, S.; Weulersse, M.; Cuisinier, M.; Stannard, K.; et al. TIGIT immune checkpoint blockade restores CD8(+) T-cell immunity against multiple myeloma. Blood 2018, 132, 1689–1694. [Google Scholar] [CrossRef]
  127. Asimakopoulos, F. TIGIT checkpoint inhibition for myeloma. Blood 2018, 132, 1629–1630. [Google Scholar] [CrossRef]
  128. Lesokhin, A.M.; Ansell, S.M.; Armand, P.; Scott, E.C.; Halwani, A.; Gutierrez, M.; Millenson, M.M.; Cohen, A.D.; Schuster, S.J.; Lebovic, D.; et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J. Clin. Oncol. 2016, 34, 2698–2704. [Google Scholar] [CrossRef]
  129. Ansell, S.; Gutierrez, M.E.; Shipp, M.A.; Gladstone, D.; Moskowitz, A.; Borello, I.; Popa-Mckiver, M.; Farsaci, B.; Zhu, L.; Lesokhin, A.M.; et al. A Phase 1 Study of Nivolumab in Combination with Ipilimumab for Relapsed or Refractory Hematologic Malignancies (CheckMate 039). Blood 2016, 128, 183. [Google Scholar] [CrossRef]
  130. Görgün, G.; Samur, M.K.; Cowens, K.B.; Paula, S.; Bianchi, G.; Anderson, J.E.; White, R.E.; Singh, A.; Ohguchi, H.; Suzuki, R.; et al. Lenalidomide Enhances Immune Checkpoint Blockade-Induced Immune Response in Multiple Myeloma. Clin. Cancer Res. 2015, 21, 4607–4618. [Google Scholar] [CrossRef]
  131. Verkleij, C.P.M.; Jhatakia, A.; Broekmans, M.E.C.; Frerichs, K.A.; Zweegman, S.; Mutis, T.; Bezman, N.A.; van de Donk, N. Preclinical Rationale for Targeting the PD-1/PD-L1 Axis in Combination with a CD38 Antibody in Multiple Myeloma and Other CD38-Positive Malignancies. Cancers 2020, 12, 3713. [Google Scholar] [CrossRef]
  132. Mateos, M.-V.; Orlowski, R.Z.; Siegel, D.S.D.; Reece, D.E.; Moreau, P.; Ocio, E.M.; Shah, J.J.; Rodríguez-Otero, P.; Munshi, N.C.; Avigan, D.; et al. Pembrolizumab in combination with lenalidomide and low-dose dexamethasone for relapsed/refractory multiple myeloma (RRMM): Final efficacy and safety analysis. J. Clin. Oncol. 2016, 34, 8010. [Google Scholar] [CrossRef]
  133. Badros, A.; Hyjek, E.; Ma, N.; Lesokhin, A.; Dogan, A.; Rapoport, A.P.; Kocoglu, M.; Lederer, E.; Philip, S.; Milliron, T.; et al. Pembrolizumab, pomalidomide, and low-dose dexamethasone for relapsed/refractory multiple myeloma. Blood 2017, 130, 1189–1197. [Google Scholar] [CrossRef]
  134. Usmani, S.Z.; Schjesvold, F.; Oriol, A.; Karlin, L.; Cavo, M.; Rifkin, R.M.; Yimer, H.A.; LeBlanc, R.; Takezako, N.; McCroskey, R.D.; et al. Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): A randomised, open-label, phase 3 trial. Lancet Haematol. 2019, 6, e448–e458. [Google Scholar] [CrossRef]
  135. Cho, H.J.; Costa, L.J.; Davies, F.E.; Neparidze, N.; Vij, R.; Feng, Y.; Teterina, A.; Wassner Fritsch, E.; Wenger, M.; Kaufman, J.L. Atezolizumab in Combination with Daratumumab with or without Lenalidomide or Pomalidomide: A Phase Ib Study in Patients with Multiple Myeloma. Blood 2018, 132, 597. [Google Scholar] [CrossRef]
  136. Verkleij, C.P.M.; Minnema, M.C.; de Weerdt, O.; Bosman, P.W.C.; Frerichs, K.A.; Croockewit, A.J.; Klein, S.K.; Bos, G.; Mutis, T.; Plattel, W.J.; et al. Efficacy and Safety of Nivolumab Combined with Daratumumab with or without Low-Dose Cyclophosphamide in Relapsed/Refractory Multiple Myeloma; Interim Analysis of the Phase 2 Nivo-Dara Study. Blood 2019, 134, 1879. [Google Scholar] [CrossRef]
  137. Cohen, Y.C.; Oriol, A.; Wu, K.L.; Lavi, N.; Vlummens, P.; Jackson, C.; Garvin, W.; Carson, R.; Crist, W.; Fu, J.; et al. Daratumumab With Cetrelimab, an Anti-PD-1 Monoclonal Antibody, in Relapsed/Refractory Multiple Myeloma. Clin. Lymphoma Myeloma Leuk 2021, 21, 46–54.e44. [Google Scholar] [CrossRef]
  138. Frerichs, K.A.; Verkleij, C.P.M.; Dimopoulos, M.A.; Marin Soto, J.A.; Zweegman, S.; Young, M.H.; Newhall, K.J.; Mutis, T.; van de Donk, N. Efficacy and Safety of Durvalumab Combined with Daratumumab in Daratumumab-Refractory Multiple Myeloma Patients. Cancers 2021, 13, 2452. [Google Scholar] [CrossRef]
  139. Tsuchikama, K.; An, Z. Antibody-drug conjugates: Recent advances in conjugation and linker chemistries. Protein Cell 2018, 9, 33–46. [Google Scholar] [CrossRef]
  140. Yu, B.; Liu, D. Antibody-drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma. J. Hematol. Oncol. 2019, 12, 94. [Google Scholar] [CrossRef]
  141. Herrera, A.F.; Molina, A. Investigational Antibody-Drug Conjugates for Treatment of B-lineage Malignancies. Clin. Lymphoma Myeloma Leuk 2018, 18, 452–468.e454. [Google Scholar] [CrossRef]
  142. Skaletskaya, A.; Setiady, Y.Y.; Park, P.U.; Lutz, R.J. Abstract 770: Lorvotuzumab mertansine (IMGN901) immune effector activity and its effect on human NK cells. Cancer Res. 2011, 71, 770. [Google Scholar] [CrossRef]
  143. Tai, Y.T.; Mayes, P.A.; Acharya, C.; Zhong, M.Y.; Cea, M.; Cagnetta, A.; Craigen, J.; Yates, J.; Gliddon, L.; Fieles, W.; et al. Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood 2014, 123, 3128–3138. [Google Scholar] [CrossRef]
  144. Yu, B.; Jiang, T.; Liu, D. BCMA-targeted immunotherapy for multiple myeloma. J. Hematol. Oncol. 2020, 13, 125. [Google Scholar] [CrossRef]
  145. Carpenter, R.O.; Evbuomwan, M.O.; Pittaluga, S.; Rose, J.J.; Raffeld, M.; Yang, S.; Gress, R.E.; Hakim, F.T.; Kochenderfer, J.N. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin. Cancer Res. 2013, 19, 2048–2060. [Google Scholar] [CrossRef]
  146. Tai, Y.T.; Anderson, K.C. Targeting B-cell maturation antigen in multiple myeloma. Immunotherapy 2015, 7, 1187–1199. [Google Scholar] [CrossRef]
  147. Zhou, J.; Wang, G.; Chen, Y.; Wang, H.; Hua, Y.; Cai, Z. Immunogenic cell death in cancer therapy: Present and emerging inducers. J. Cell. Mol. Med. 2019, 23, 4854–4865. [Google Scholar] [CrossRef]
  148. Trudel, S.; Lendvai, N.; Popat, R.; Voorhees, P.M.; Reeves, B.; Libby, E.N.; Richardson, P.G.; Anderson, L.D., Jr.; Sutherland, H.J.; Yong, K.; et al. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma (BMA117159): A dose escalation and expansion phase 1 trial. Lancet Oncol. 2018, 19, 1641–1653. [Google Scholar] [CrossRef]
  149. Lonial, S.; Lee, H.C.; Badros, A.; Trudel, S.; Nooka, A.K.; Chari, A.; Abdallah, A.O.; Callander, N.; Lendvai, N.; Sborov, D.; et al. Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): A two-arm, randomised, open-label, phase 2 study. Lancet Oncol. 2020, 21, 207–221. [Google Scholar] [CrossRef]
  150. Trudel, S.; Lendvai, N.; Popat, R.; Voorhees, P.M.; Reeves, B.; Libby, E.N.; Richardson, P.G.; Hoos, A.; Gupta, I.; Bragulat, V.; et al. Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: An update on safety and efficacy from dose expansion phase I study. Blood Cancer J. 2019, 9, 37. [Google Scholar] [CrossRef]
  151. Richardson, P.G.; Lee, H.C.; Abdallah, A.O.; Cohen, A.D.; Kapoor, P.; Voorhees, P.M.; Hoos, A.; Wang, K.; Baron, J.; Piontek, T.; et al. Single-agent belantamab mafodotin for relapsed/refractory multiple myeloma: Analysis of the lyophilised presentation cohort from the pivotal DREAMM-2 study. Blood Cancer J. 2020, 10, 106. [Google Scholar] [CrossRef]
  152. Lonial, S.; Lee, H.C.; Badros, A.; Trudel, S.; Nooka, A.K.; Chari, A.; Abdallah, A.O.; Callander, N.; Sborov, D.; Suvannasankha, A.; et al. Longer term outcomes with single-agent belantamab mafodotin in patients with relapsed or refractory multiple myeloma: 13-month follow-up from the pivotal DREAMM-2 study. Cancer 2021, 127, 4198–4212. [Google Scholar] [CrossRef]
  153. Tai, Y.T.; Anderson, K.C. B cell maturation antigen (BCMA)-based immunotherapy for multiple myeloma. Expert Opin. Biol. Ther. 2019, 19, 1143–1156. [Google Scholar] [CrossRef]
  154. Lee, H.C.; Raje, N.S.; Landgren, O.; Upreti, V.V.; Wang, J.; Avilion, A.A.; Hu, X.; Rasmussen, E.; Ngarmchamnanrith, G.; Fujii, H.; et al. Phase 1 study of the anti-BCMA antibody-drug conjugate AMG 224 in patients with relapsed/refractory multiple myeloma. Leukemia 2021, 35, 255–258. [Google Scholar] [CrossRef]
  155. Kinneer, K.; Flynn, M.; Thomas, S.B.; Meekin, J.; Varkey, R.; Xiao, X.; Zhong, H.; Breen, S.; Hynes, P.G.; Fleming, R.; et al. Preclinical assessment of an antibody-PBD conjugate that targets BCMA on multiple myeloma and myeloma progenitor cells. Leukemia 2019, 33, 766–771. [Google Scholar] [CrossRef]
  156. Xing, L.; Lin, L.; Yu, T.; Li, Y.; Wen, K.; Cho, S.-F.; Hsieh, P.A.; Kinneer, K.; Munshi, N.C.; Anderson, K.C.; et al. Anti-Bcma PBD MEDI2228 Combats Drug Resistance and Synergizes with Bortezomib and Inhibitors to DNA Damage Response in Multiple Myeloma. Blood 2019, 134, 1817. [Google Scholar] [CrossRef]
  157. Xing, L.; Lin, L.; Yu, T.; Li, Y.; Cho, S.F.; Liu, J.; Wen, K.; Hsieh, P.A.; Kinneer, K.; Munshi, N.; et al. A novel BCMA PBD-ADC with ATM/ATR/WEE1 inhibitors or bortezomib induce synergistic lethality in multiple myeloma. Leukemia 2020, 34, 2150–2162. [Google Scholar] [CrossRef]
  158. Kumar, S.K.; Migkou, M.; Bhutani, M.; Spencer, A.; Ailawadhi, S.; Kalff, A.; Walcott, F.; Pore, N.; Gibson, D.; Wang, F.; et al. Phase 1, First-in-Human Study of MEDI2228, a BCMA-Targeted ADC in Patients with Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 26–27. [Google Scholar] [CrossRef]
  159. Willert, E.K.; Robinson, G.L.; Higgins, J.P.; Liu, J.; Lee, J.; Syed, S.; Zhang, Y.; Tavares, D.; Lublinsky, A.; Chattopadhyay, N.; et al. Abstract 2384: TAK-169, an exceptionally potent CD38 targeted engineered toxin body, as a novel direct cell kill approach for the treatment of multiple myeloma. Cancer Res. 2019, 79, 2384. [Google Scholar] [CrossRef]
  160. Vogl, D.T.; Kaufman, J.L.; Holstein, S.A.; Nadeem, O.; O’Donnell, E.; Suryanarayan, K.; Collins, S.; Parot, X.; Chaudhry, M. TAK-573, an Anti-CD38/Attenuated Ifnα Fusion Protein, Has Clinical Activity and Modulates the Ifnα Receptor (IFNAR) Pathway in Patients with Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 37–38. [Google Scholar] [CrossRef]
  161. Sherbenou, D.W.; Aftab, B.T.; Su, Y.; Behrens, C.R.; Wiita, A.; Logan, A.C.; Acosta-Alvear, D.; Hann, B.C.; Walter, P.; Shuman, M.A.; et al. Antibody-drug conjugate targeting CD46 eliminates multiple myeloma cells. J. Clin. Investig. 2016, 126, 4640–4653. [Google Scholar] [CrossRef]
  162. Burton, J.D.; Ely, S.; Reddy, P.K.; Stein, R.; Gold, D.V.; Cardillo, T.M.; Goldenberg, D.M. CD74 is expressed by multiple myeloma and is a promising target for therapy. Clin. Cancer Res. 2004, 10, 6606–6611. [Google Scholar] [CrossRef]
  163. Stein, R.; Mattes, M.J.; Cardillo, T.M.; Hansen, H.J.; Chang, C.H.; Burton, J.; Govindan, S.; Goldenberg, D.M. CD74: A new candidate target for the immunotherapy of B-cell neoplasms. Clin. Cancer Res. 2007, 13, 5556s–5563s. [Google Scholar] [CrossRef]
  164. Abrahams, C.L.; Li, X.; Embry, M.; Yu, A.; Krimm, S.; Krueger, S.; Greenland, N.Y.; Wen, K.W.; Jones, C.; DeAlmeida, V.; et al. Targeting CD74 in multiple myeloma with the novel, site-specific antibody-drug conjugate STRO-001. Oncotarget 2018, 9, 37700–37714. [Google Scholar] [CrossRef]
  165. Shah, N.N.; Krishnan, A.Y.; Shah, N.D.; Burke, J.M.; Melear, J.M.; Spira, A.I.; Popplewell, L.L.; Andreadis, C.B.; Chhabra, S.; Sharman, J.P.; et al. Preliminary Results of a Phase 1 Dose Escalation Study of the First-in-Class Anti-CD74 Antibody Drug Conjugate (ADC), STRO-001, in Patients with Advanced B-Cell Malignancies. Blood 2019, 134, 5329. [Google Scholar] [CrossRef]
  166. Kontermann, R.E.; Brinkmann, U. Bispecific antibodies. Drug Discov. Today 2015, 20, 838–847. [Google Scholar] [CrossRef]
  167. Lancman, G.; Sastow, D.L.; Cho, H.J.; Jagannath, S.; Madduri, D.; Parekh, S.S.; Richard, S.; Richter, J.; Sanchez, L.; Chari, A. Bispecific Antibodies in Multiple Myeloma: Present and Future. Blood Cancer Discov. 2021, 2, 423–433. [Google Scholar] [CrossRef]
  168. Smith, E.L.; Harrington, K.; Staehr, M.; Masakayan, R.; Jones, J.; Long, T.J.; Ng, K.Y.; Ghoddusi, M.; Purdon, T.J.; Wang, X.; et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci. Transl. Med. 2019, 11, eaau7746. [Google Scholar] [CrossRef]
  169. Elkins, K.; Zheng, B.; Go, M.; Slaga, D.; Du, C.; Scales, S.J.; Yu, S.F.; McBride, J.; de Tute, R.; Rawstron, A.; et al. FcRL5 as a target of antibody-drug conjugates for the treatment of multiple myeloma. Mol. Cancer Ther. 2012, 11, 2222–2232. [Google Scholar] [CrossRef]
  170. Dement-Brown, J.; Newton, C.S.; Ise, T.; Damdinsuren, B.; Nagata, S.; Tolnay, M. Fc receptor-like 5 promotes B cell proliferation and drives the development of cells displaying switched isotypes. J. Leukoc. Biol. 2012, 91, 59–67. [Google Scholar] [CrossRef]
  171. Ross, T.; Reusch, U.; Wingert, S.; Haneke, T.; Klausz, K.; Otte, A.-K.; Schub, N.; Knackmuss, S.; Müller, T.; Ellwanger, K.; et al. Preclinical Characterization of AFM26, a Novel B Cell Maturation Antigen (BCMA)-Directed Tetravalent Bispecific Antibody for High Affinity Retargeting of NK Cells Against Myeloma. Blood 2018, 132, 1927. [Google Scholar] [CrossRef]
  172. Draghi, M.; Schafer, J.L.; Nelson, A.; Frye, Z.; Oliphant, A.; Haserlat, S.; Lajoie, J.; Rogers, K.; Villinger, F.; Schmidt, M.; et al. Abstract 4972: Preclinical development of a first-in-class NKp30xBCMA NK cell engager for the treatment of multiple myeloma. Cancer Res. 2019, 79, 4972. [Google Scholar] [CrossRef]
  173. Watkins-Yoon, J.; Guzman, W.; Oliphant, A.; Haserlat, S.; Leung, A.; Chottin, C.; Ophir, M.; Vekeria, J.; Nelson, A.P.; Frye, Z.; et al. CTX-8573, an Innate-Cell Engager Targeting BCMA, is a Highly Potent Multispecific Antibody for the Treatment of Multiple Myeloma. Blood 2019, 134, 3182. [Google Scholar] [CrossRef]
  174. Shah, Z.; Malik, M.N.; Batool, S.S.; Kotapati, S.; Akhtar, A.; Rehman, O.u.; Ghani, M.; Sadiq, M.; Akbar, A.; Ashraf, A.; et al. Bispecific T-Cell Engager (BiTE) Antibody Based Immunotherapy for Treatment of Relapsed Refractory Multiple Myeloma (RRMM): A Systematic Review of Preclinical and Clinical Trials. Blood 2019, 134, 5567. [Google Scholar] [CrossRef]
  175. Harrison, S.J.; Minnema, M.C.; Lee, H.C.; Spencer, A.; Kapoor, P.; Madduri, D.; Larsen, J.; Ailawadhi, S.; Kaufman, J.L.; Raab, M.S.; et al. A Phase 1 First in Human (FIH) Study of AMG 701, an Anti-B-Cell Maturation Antigen (BCMA) Half-Life Extended (HLE) BiTE® (bispecific T-cell engager) Molecule, in Relapsed/Refractory (RR) Multiple Myeloma (MM). Blood 2020, 136, 28–29. [Google Scholar] [CrossRef]
  176. Cho, S.F.; Lin, L.; Xing, L.; Li, Y.; Wen, K.; Yu, T.; Hsieh, P.A.; Munshi, N.; Wahl, J.; Matthes, K.; et al. The immunomodulatory drugs lenalidomide and pomalidomide enhance the potency of AMG 701 in multiple myeloma preclinical models. Blood Adv. 2020, 4, 4195–4207. [Google Scholar] [CrossRef]
  177. Hipp, S.; Tai, Y.T.; Blanset, D.; Deegen, P.; Wahl, J.; Thomas, O.; Rattel, B.; Adam, P.J.; Anderson, K.C.; Friedrich, M. A novel BCMA/CD3 bispecific T-cell engager for the treatment of multiple myeloma induces selective lysis in vitro and in vivo. Leukemia 2017, 31, 1743–1751. [Google Scholar] [CrossRef]
  178. Costa, L.J.; Wong, S.W.; Bermúdez, A.; de la Rubia, J.; Mateos, M.-V.; Ocio, E.M.; Rodríguez-Otero, P.; San-Miguel, J.; Li, S.; Sarmiento, R.; et al. First Clinical Study of the B-Cell Maturation Antigen (BCMA) 2+1 T Cell Engager (TCE) CC-93269 in Patients (Pts) with Relapsed/Refractory Multiple Myeloma (RRMM): Interim Results of a Phase 1 Multicenter Trial. Blood 2019, 134, 143. [Google Scholar] [CrossRef]
  179. Chari, A.; Berdeja, J.G.; Oriol, A.; van de Donk, N.W.C.J.; Rodriguez, P.; Askari, E.; Mateos, M.-V.; Minnema, M.C.; Verona, R.; Girgis, S.; et al. A Phase 1, First-in-Human Study of Talquetamab, a G Protein-Coupled Receptor Family C Group 5 Member D (GPRC5D) × CD3 Bispecific Antibody, in Patients with Relapsed and/or Refractory Multiple Myeloma (RRMM). Blood 2020, 136, 40–41. [Google Scholar] [CrossRef]
  180. Girgis, S.; Lin, S.X.W.; Pillarisetti, K.; Verona, R.; Vieyra, D.; Casneuf, T.; Fink, D.; Miao, X.; Chen, Y.; Stephenson, T.; et al. Teclistamab and talquetamab modulate levels of soluble B-cell maturation antigen in patients with relapsed and/or refractory multiple myeloma. J. Clin. Oncol. 2021, 39, 8047. [Google Scholar] [CrossRef]
  181. Berdeja, J.G.; Krishnan, A.Y.; Oriol, A.; Donk, N.W.C.J.v.d.; Rodríguez-Otero, P.; Askari, E.; Mateos, M.-V.; Minnema, M.C.; Costa, L.J.; Verona, R.; et al. Updated results of a phase 1, first-in-human study of talquetamab, a G protein-coupled receptor family C group 5 member D (GPRC5D) × CD3 bispecific antibody, in relapsed/refractory multiple myeloma (MM). J. Clin. Oncol. 2021, 39, 8008. [Google Scholar] [CrossRef]
  182. Rodriguez, C.; D’Souza, A.; Shah, N.; Voorhees, P.M.; Buelow, B.; Vij, R.; Kumar, S.K. Initial Results of a Phase I Study of TNB-383B, a BCMA × CD3 Bispecific T-Cell Redirecting Antibody, in Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 43–44. [Google Scholar] [CrossRef]
  183. Cohen, A.D.; Harrison, S.J.; Krishnan, A.; Fonseca, R.; Forsberg, P.A.; Spencer, A.; Berdeja, J.G.; Laubach, J.P.; Li, M.; Choeurng, V.; et al. Initial Clinical Activity and Safety of BFCR4350A, a FcRH5/CD3 T-Cell-Engaging Bispecific Antibody, in Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 42–43. [Google Scholar] [CrossRef]
  184. Wu, L.; Seung, E.; Xu, L.; Rao, E.; Lord, D.M.; Wei, R.R.; Cortez-Retamozo, V.; Ospina, B.; Posternak, V.; Ulinski, G.; et al. Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation. Nat. Cancer 2020, 1, 86–98. [Google Scholar] [CrossRef]
  185. Lancman, G.; Richter, J.; Chari, A. Bispecifics, trispecifics, and other novel immune treatments in myeloma. Hematol. Am. Soc. Hematol. Educ. Program 2020, 2020, 264–271. [Google Scholar] [CrossRef]
  186. Plesner, T.; Harrison, S.J.; Quach, H.; Lee, C.H.; Bryant, A.; Vangsted, A.J.; Estell, J.; Delforge, M.; Offner, F.; Twomey, P.; et al. A Phase I Study of RO7297089, a B-Cell Maturation Antigen (BCMA)-CD16a Bispecific Antibody in Patients with Relapsed/Refractory Multiple Myeloma (RRMM). Blood 2021, 138, 2755. [Google Scholar] [CrossRef]
  187. Chan, W.K.; Kang, S.; Youssef, Y.; Glankler, E.N.; Barrett, E.R.; Carter, A.M.; Ahmed, E.H.; Prasad, A.; Chen, L.; Zhang, J.; et al. A CS1-NKG2D Bispecific Antibody Collectively Activates Cytolytic Immune Cells against Multiple Myeloma. Cancer Immunol. Res. 2018, 6, 776–787. [Google Scholar] [CrossRef]
  188. Gantke, T.; Weichel, M.; Herbrecht, C.; Reusch, U.; Ellwanger, K.; Fucek, I.; Eser, M.; Müller, T.; Griep, R.; Molkenthin, V.; et al. Trispecific antibodies for CD16A-directed NK cell engagement and dual-targeting of tumor cells. Protein Eng. Des. Sel. 2017, 30, 673–684. [Google Scholar] [CrossRef]
  189. Sadelain, M.; Brentjens, R.; Rivière, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013, 3, 388–398. [Google Scholar] [CrossRef]
  190. Turtle, C.J.; Hudecek, M.; Jensen, M.C.; Riddell, S.R. Engineered T cells for anti-cancer therapy. Curr. Opin Immunol. 2012, 24, 633–639. [Google Scholar] [CrossRef]
  191. Sadelain, M.; Rivière, I.; Riddell, S. Therapeutic T cell engineering. Nature 2017, 545, 423–431. [Google Scholar] [CrossRef] [PubMed]
  192. 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]
  193. Brudno, J.N.; Maric, I.; Hartman, S.D.; Rose, J.J.; Wang, M.; Lam, N.; Stetler-Stevenson, M.; Salem, D.; Yuan, C.; Pavletic, S.; et al. T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J. Clin. Oncol. 2018, 36, 2267–2280. [Google Scholar] [CrossRef] [PubMed]
  194. Till, B.G.; Jensen, M.C.; Wang, J.; Chen, E.Y.; Wood, B.L.; Greisman, H.A.; Qian, X.; James, S.E.; Raubitschek, A.; Forman, S.J.; et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 2008, 112, 2261–2271. [Google Scholar] [CrossRef] [PubMed]
  195. Mikkilineni, L.; Kochenderfer, J.N. Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood 2017, 130, 2594–2602. [Google Scholar] [CrossRef]
  196. Lim, W.A.; June, C.H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168, 724–740. [Google Scholar] [CrossRef]
  197. Levine, B.L.; Miskin, J.; Wonnacott, K.; Keir, C. Global Manufacturing of CAR T Cell Therapy. Mol. Ther. Methods Clin. Dev. 2017, 4, 92–101. [Google Scholar] [CrossRef]
  198. Srivastava, S.; Riddell, S.R. Engineering CAR-T cells: Design concepts. Trends Immunol. 2015, 36, 494–502. [Google Scholar] [CrossRef]
  199. Wang, X.; Rivière, I. Clinical manufacturing of CAR T cells: Foundation of a promising therapy. Mol. Ther. Oncolytics 2016, 3, 16015. [Google Scholar] [CrossRef]
  200. Gattinoni, L.; Finkelstein, S.E.; Klebanoff, C.A.; Antony, P.A.; Palmer, D.C.; Spiess, P.J.; Hwang, L.N.; Yu, Z.; Wrzesinski, C.; Heimann, D.M.; et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 2005, 202, 907–912. [Google Scholar] [CrossRef]
  201. Cohen, A.D.; Garfall, A.L.; Stadtmauer, E.A.; Melenhorst, J.J.; Lacey, S.F.; Lancaster, E.; Vogl, D.T.; Weiss, B.M.; Dengel, K.; Nelson, A.; et al. B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma. J. Clin. Investig. 2019, 129, 2210–2221. [Google Scholar] [CrossRef] [PubMed]
  202. 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]
  203. 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]
  204. Friedman, K.M.; Garrett, T.E.; Evans, J.W.; Horton, H.M.; Latimer, H.J.; Seidel, S.L.; Horvath, C.J.; Morgan, R.A. Effective Targeting of Multiple B-Cell Maturation Antigen-Expressing Hematological Malignances by Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T Cells. Hum. Gene Ther. 2018, 29, 585–601. [Google Scholar] [CrossRef]
  205. 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]
  206. 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]
  207. Xu, J.; Chen, L.J.; Yang, S.S.; Sun, Y.; Wu, W.; Liu, Y.F.; Xu, J.; Zhuang, Y.; Zhang, W.; Weng, X.Q.; et al. Exploratory trial of a biepitopic CAR T-targeting B cell maturation antigen in relapsed/refractory multiple myeloma. Proc. Natl. Acad. Sci. USA 2019, 116, 9543–9551. [Google Scholar] [CrossRef]
  208. Van de Donk, N.; Themeli, M.; Usmani, S.Z. Determinants of response and mechanisms of resistance of CAR T-cell therapy in multiple myeloma. Blood Cancer Discov. 2021, 2, 302–318. [Google Scholar] [CrossRef]
  209. Li, C.; Wang, Q.; Zhu, H.; Mao, X.; Wang, Y.; Zhang, Y.; Zhou, J. T Cells Expressing Anti B-Cell Maturation Antigen Chimeric Antigen Receptors for Plasma Cell Malignancies. Blood 2018, 132, 1013. [Google Scholar] [CrossRef]
  210. Pont, M.J.; Hill, T.; Cole, G.O.; Abbott, J.J.; Kelliher, J.; Salter, A.I.; Hudecek, M.; Comstock, M.L.; Rajan, A.; Patel, B.K.R.; et al. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 2019, 134, 1585–1597. [Google Scholar] [CrossRef]
  211. Green, D.J.; Pont, M.; Sather, B.D.; Cowan, A.J.; Turtle, C.J.; Till, B.G.; Nagengast, A.M.; Libby, E.N., III; Becker, P.S.; Coffey, D.G.; et al. Fully Human Bcma Targeted Chimeric Antigen Receptor T Cells Administered in a Defined Composition Demonstrate Potency at Low Doses in Advanced Stage High Risk Multiple Myeloma. Blood 2018, 132, 1011. [Google Scholar] [CrossRef]
  212. Samur, M.K.; Fulciniti, M.; Aktas Samur, A.; Bazarbachi, A.H.; Tai, Y.T.; Prabhala, R.; Alonso, A.; Sperling, A.S.; Campbell, T.; Petrocca, F.; et al. Biallelic loss of BCMA as a resistance mechanism to CAR T cell therapy in a patient with multiple myeloma. Nat. Commun. 2021, 12, 868. [Google Scholar] [CrossRef] [PubMed]
  213. Da Vià, M.C.; Dietrich, O.; Truger, M.; Arampatzi, P.; Duell, J.; Heidemeier, A.; Zhou, X.; Danhof, S.; Kraus, S.; Chatterjee, M.; et al. Homozygous BCMA gene deletion in response to anti-BCMA CAR T cells in a patient with multiple myeloma. Nat. Med. 2021, 27, 616–619. [Google Scholar] [CrossRef] [PubMed]
  214. Long, A.H.; Haso, W.M.; Shern, J.F.; Wanhainen, K.M.; Murgai, M.; Ingaramo, M.; Smith, J.P.; Walker, A.J.; Kohler, M.E.; Venkateshwara, V.R.; et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015, 21, 581–590. [Google Scholar] [CrossRef] [PubMed]
  215. Zah, E.; Nam, E.; Bhuvan, V.; Tran, U.; Ji, B.Y.; Gosliner, S.B.; Wang, X.; Brown, C.E.; Chen, Y.Y. Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma. Nat. Commun. 2020, 11, 2283. [Google Scholar] [CrossRef]
  216. Cherkassky, L.; Morello, A.; Villena-Vargas, J.; Feng, Y.; Dimitrov, D.S.; Jones, D.R.; Sadelain, M.; Adusumilli, P.S. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Investig. 2016, 126, 3130–3144. [Google Scholar] [CrossRef]
  217. Medema, J.P.; de Jong, J.; van Hall, T.; Melief, C.J.; Offringa, R. Immune escape of tumors in vivo by expression of cellular FLICE-inhibitory protein. J. Exp. Med. 1999, 190, 1033–1038. [Google Scholar] [CrossRef]
  218. Medema, J.P.; de Jong, J.; Peltenburg, L.T.; Verdegaal, E.M.; Gorter, A.; Bres, S.A.; Franken, K.L.; Hahne, M.; Albar, J.P.; Melief, C.J.; et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 11515–11520. [Google Scholar] [CrossRef]
  219. Wang, M.; Pruteanu, I.; Cohen, A.D.; Garfall, A.L.; Milone, M.C.; Tian, L.; Gonzalez, V.E.; Gill, S.; Frey, N.V.; Barrett, D.M.; et al. Identification and Validation of Predictive Biomarkers to CD19- and BCMA-Specific CAR T-Cell Responses in CAR T-Cell Precursors. Blood 2019, 134, 622. [Google Scholar] [CrossRef]
  220. Finney, O.C.; Yeri, A.; Mao, P.; Pandya, C.; Alonzo, E.; Hopkins, G.; Hymson, S.; Hu, T.; Foos, M.; Bhadoriya, S.; et al. Molecular and Phenotypic Profiling of Drug Product and Post-Infusion Samples from CRB-402, an Ongoing: Phase I Clinical Study of bb21217 a BCMA-Directed CAR T Cell Therapy. Blood 2020, 136, 3–4. [Google Scholar] [CrossRef]
  221. Leblay, N.; Maity, R.; Barakat, E.; McCulloch, S.; Duggan, P.; Jimenez-Zepeda, V.; Bahlis, N.J.; Neri, P. Cite-Seq Profiling of T Cells in Multiple Myeloma Patients Undergoing BCMA Targeting CAR-T or Bites Immunotherapy. Blood 2020, 136, 11–12. [Google Scholar] [CrossRef]
  222. Garfall, A.L.; Dancy, E.K.; Cohen, A.D.; Hwang, W.T.; Fraietta, J.A.; Davis, M.M.; Levine, B.L.; Siegel, D.L.; Stadtmauer, E.A.; Vogl, D.T.; et al. T-cell phenotypes associated with effective CAR T-cell therapy in postinduction vs relapsed multiple myeloma. Blood Adv. 2019, 3, 2812–2815. [Google Scholar] [CrossRef] [PubMed]
  223. Sommermeyer, D.; Hudecek, M.; Kosasih, P.L.; Gogishvili, T.; Maloney, D.G.; Turtle, C.J.; Riddell, S.R. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016, 30, 492–500. [Google Scholar] [CrossRef] [PubMed]
  224. Adusumilli, P.S.; Cherkassky, L.; Villena-Vargas, J.; Colovos, C.; Servais, E.; Plotkin, J.; Jones, D.R.; Sadelain, M. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci. Transl. Med. 2014, 6, 261ra151. [Google Scholar] [CrossRef]
  225. Ruella, M.; Barrett, D.M.; Kenderian, S.S.; Shestova, O.; Hofmann, T.J.; Perazzelli, J.; Klichinsky, M.; Aikawa, V.; Nazimuddin, F.; Kozlowski, M.; et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J. Clin. Investig. 2016, 126, 3814–3826. [Google Scholar] [CrossRef]
  226. Chen, K.H.; Wada, M.; Pinz, K.G.; Liu, H.; Shuai, X.; Chen, X.; Yan, L.E.; Petrov, J.C.; Salman, H.; Senzel, L.; et al. A compound chimeric antigen receptor strategy for targeting multiple myeloma. Leukemia 2018, 32, 402–412. [Google Scholar] [CrossRef]
  227. Fernández de Larrea, C.; Staehr, M.; Lopez, A.V.; Ng, K.Y.; Chen, Y.; Godfrey, W.D.; Purdon, T.J.; Ponomarev, V.; Wendel, H.G.; Brentjens, R.J.; et al. Defining an Optimal Dual-Targeted CAR T-cell Therapy Approach Simultaneously Targeting BCMA and GPRC5D to Prevent BCMA Escape-Driven Relapse in Multiple Myeloma. Blood Cancer Discov. 2020, 1, 146–154. [Google Scholar] [CrossRef]
  228. Suarez, E.R.; de Chang, K.; Sun, J.; Sui, J.; Freeman, G.J.; Signoretti, S.; Zhu, Q.; Marasco, W.A. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget 2016, 7, 34341–34355. [Google Scholar] [CrossRef]
  229. Li, S.; Siriwon, N.; Zhang, X.; Yang, S.; Jin, T.; He, F.; Kim, Y.J.; Mac, J.; Lu, Z.; Wang, S.; et al. Enhanced Cancer Immunotherapy by Chimeric Antigen Receptor-Modified T Cells Engineered to Secrete Checkpoint Inhibitors. Clin. Cancer Res. 2017, 23, 6982–6992. [Google Scholar] [CrossRef]
  230. Gargett, T.; Yu, W.; Dotti, G.; Yvon, E.S.; Christo, S.N.; Hayball, J.D.; Lewis, I.D.; Brenner, M.K.; Brown, M.P. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade. Mol. Ther. 2016, 24, 1135–1149. [Google Scholar] [CrossRef]
  231. Drent, E.; Poels, R.; Ruiter, R.; van de Donk, N.; Zweegman, S.; Yuan, H.; de Bruijn, J.; Sadelain, M.; Lokhorst, H.M.; Groen, R.W.J.; et al. Combined CD28 and 4-1BB Costimulation Potentiates Affinity-tuned Chimeric Antigen Receptor-engineered T Cells. Clin. Cancer Res. 2019, 25, 4014–4025. [Google Scholar] [CrossRef] [PubMed]
  232. Zhao, Z.; Condomines, M.; van der Stegen, S.J.C.; Perna, F.; Kloss, C.C.; Gunset, G.; Plotkin, J.; Sadelain, M. Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T Cells. Cancer Cell 2015, 28, 415–428. [Google Scholar] [CrossRef] [PubMed]
  233. Mailankody, S.; Matous, J.V.; Liedtke, M.; Sidana, S.; Malik, S.; Nath, R.; Oluwole, O.O.; Karski, E.E.; Lovelace, W.; Zhou, X.; et al. Universal: An Allogeneic First-in-Human Study of the Anti-Bcma ALLO-715 and the Anti-CD52 ALLO-647 in Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 24–25. [Google Scholar] [CrossRef]
  234. Carmon, L.; Avivi, I.; Kovjazin, R.; Zuckerman, T.; Dray, L.; Gatt, M.E.; Or, R.; Shapira, M.Y. Phase I/II study exploring ImMucin, a pan-major histocompatibility complex, anti-MUC1 signal peptide vaccine, in multiple myeloma patients. Br. J. Haematol. 2015, 169, 44–56. [Google Scholar] [CrossRef]
  235. Kovjazin, R.; Volovitz, I.; Kundel, Y.; Rosenbaum, E.; Medalia, G.; Horn, G.; Smorodinsky, N.I.; Brenner, B.; Carmon, L. ImMucin: A novel therapeutic vaccine with promiscuous MHC binding for the treatment of MUC1-expressing tumors. Vaccine 2011, 29, 4676–4686. [Google Scholar] [CrossRef] [PubMed]
  236. Kovjazin, R.; Horn, G.; Smorodinsky, N.I.; Shapira, M.Y.; Carmon, L. Cell surface-associated anti-MUC1-derived signal peptide antibodies: Implications for cancer diagnostics and therapy. PLoS ONE 2014, 9, e85400. [Google Scholar] [CrossRef]
  237. Choi, C.; Witzens, M.; Bucur, M.; Feuerer, M.; Sommerfeldt, N.; Trojan, A.; Ho, A.; Schirrmacher, V.; Goldschmidt, H.; Beckhove, P. Enrichment of functional CD8 memory T cells specific for MUC1 in bone marrow of patients with multiple myeloma. Blood 2005, 105, 2132–2134. [Google Scholar] [CrossRef]
  238. Bae, J.; Samur, M.; Munshi, A.; Hideshima, T.; Keskin, D.; Kimmelman, A.; Lee, A.H.; Dranoff, G.; Anderson, K.C.; Munshi, N.C. Heteroclitic XBP1 peptides evoke tumor-specific memory cytotoxic T lymphocytes against breast cancer, colon cancer, and pancreatic cancer cells. Oncoimmunology 2014, 3, e970914. [Google Scholar] [CrossRef]
  239. Bae, J.; Prabhala, R.; Voskertchian, A.; Brown, A.; Maguire, C.; Richardson, P.; Dranoff, G.; Anderson, K.C.; Munshi, N.C. A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients. Leukemia 2015, 29, 218–229. [Google Scholar] [CrossRef]
  240. Nooka, A.K.; Wang, M.L.; Yee, A.J.; Kaufman, J.L.; Bae, J.; Peterkin, D.; Richardson, P.G.; Raje, N.S. Assessment of Safety and Immunogenicity of PVX-410 Vaccine With or Without Lenalidomide in Patients With Smoldering Multiple Myeloma: A Nonrandomized Clinical Trial. JAMA Oncol. 2018, 4, e183267. [Google Scholar] [CrossRef]
  241. Jørgensen, N.G.; Klausen, U.; Grauslund, J.H.; Helleberg, C.; Aagaard, T.G.; Do, T.H.; Ahmad, S.M.; Olsen, L.R.; Klausen, T.W.; Breinholt, M.F.; et al. Peptide Vaccination Against PD-L1 With IO103 a Novel Immune Modulatory Vaccine in Multiple Myeloma: A Phase I First-in-Human Trial. Front. Immunol. 2020, 11, 595035. [Google Scholar] [CrossRef] [PubMed]
  242. Hallett, W.H.; Jing, W.; Drobyski, W.R.; Johnson, B.D. Immunosuppressive effects of multiple myeloma are overcome by PD-L1 blockade. Biol. Blood Marrow Transplant. 2011, 17, 1133–1145. [Google Scholar] [CrossRef] [PubMed]
  243. Rosenblatt, J.; Avivi, I.; Vasir, B.; Uhl, L.; Munshi, N.C.; Katz, T.; Dey, B.R.; Somaiya, P.; Mills, H.; Campigotto, F.; et al. Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin. Cancer Res. 2013, 19, 3640–3648. [Google Scholar] [CrossRef] [PubMed]
  244. Billel, G.; Smith, E.L.; Dogan, A.; Hsu, M.; Devlin, S.; Pichardo, J.D.; Chung, D.J.; Koehne, G.; Korde, N.S.; Landau, H.J.; et al. Presence of PD-1 Expressing T Cells Predicts for Inferior Overall Survival in Newly Diagnosed Multiple Myeloma. Blood 2015, 126, 1785. [Google Scholar] [CrossRef]
  245. Qian, J.; Xie, J.; Hong, S.; Yang, J.; Zhang, L.; Han, X.; Wang, M.; Zhan, F.; Shaughnessy, J.D., Jr.; Epstein, J.; et al. Dickkopf-1 (DKK1) is a widely expressed and potent tumor-associated antigen in multiple myeloma. Blood 2007, 110, 1587–1594. [Google Scholar] [CrossRef]
  246. Qian, J.; Zheng, Y.; Zheng, C.; Wang, L.; Qin, H.; Hong, S.; Li, H.; Lu, Y.; He, J.; Yang, J.; et al. Active vaccination with Dickkopf-1 induces protective and therapeutic antitumor immunity in murine multiple myeloma. Blood 2012, 119, 161–169. [Google Scholar] [CrossRef]
  247. Strambio-De-Castillia, C.; Niepel, M.; Rout, M.P. The nuclear pore complex: Bridging nuclear transport and gene regulation. Nat. Rev. Mol. Cell Biol. 2010, 11, 490–501. [Google Scholar] [CrossRef]
  248. Theodoropoulos, N.; Lancman, G.; Chari, A. Targeting Nuclear Export Proteins in Multiple Myeloma Therapy. Target. Oncol. 2020, 15, 697–708. [Google Scholar] [CrossRef]
  249. Azizian, N.G.; Li, Y. XPO1-dependent nuclear export as a target for cancer therapy. J. Hematol. Oncol. 2020, 13, 61. [Google Scholar] [CrossRef]
  250. Turner, J.G.; Dawson, J.; Sullivan, D.M. Nuclear export of proteins and drug resistance in cancer. Biochem. Pharmacol. 2012, 83, 1021–1032. [Google Scholar] [CrossRef]
  251. Schmidt, J.; Braggio, E.; Kortuem, K.M.; Egan, J.B.; Zhu, Y.X.; Xin, C.S.; Tiedemann, R.E.; Palmer, S.E.; Garbitt, V.M.; McCauley, D.; et al. Genome-wide studies in multiple myeloma identify XPO1/CRM1 as a critical target validated using the selective nuclear export inhibitor KPT-276. Leukemia 2013, 27, 2357–2365. [Google Scholar] [CrossRef] [PubMed]
  252. Kashyap, T.; Argueta, C.; Aboukameel, A.; Unger, T.J.; Klebanov, B.; Mohammad, R.M.; Muqbil, I.; Azmi, A.S.; Drolen, C.; Senapedis, W.; et al. Selinexor, a Selective Inhibitor of Nuclear Export (SINE) compound, acts through NF-κB deactivation and combines with proteasome inhibitors to synergistically induce tumor cell death. Oncotarget 2016, 7, 78883–78895. [Google Scholar] [CrossRef] [PubMed]
  253. Turner, J.G.; Kashyap, T.; Dawson, J.L.; Gomez, J.; Bauer, A.A.; Grant, S.; Dai, Y.; Shain, K.H.; Meads, M.; Landesman, Y.; et al. XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IκBα and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget 2016, 7, 78896–78909. [Google Scholar] [CrossRef] [PubMed]
  254. Gandhi, U.H.; Senapedis, W.; Baloglu, E.; Unger, T.J.; Chari, A.; Vogl, D.; Cornell, R.F. Clinical Implications of Targeting XPO1-mediated Nuclear Export in Multiple Myeloma. Clin. Lymphoma Myeloma Leuk 2018, 18, 335–345. [Google Scholar] [CrossRef]
  255. Kanai, M.; Hanashiro, K.; Kim, S.H.; Hanai, S.; Boulares, A.H.; Miwa, M.; Fukasawa, K. Inhibition of Crm1-p53 interaction and nuclear export of p53 by poly(ADP-ribosyl)ation. Nat. Cell Biol. 2007, 9, 1175–1183. [Google Scholar] [CrossRef] [PubMed]
  256. Vogt, P.K.; Jiang, H.; Aoki, M. Triple layer control: Phosphorylation, acetylation and ubiquitination of FOXO proteins. Cell Cycle 2005, 4, 908–913. [Google Scholar] [CrossRef]
  257. Tai, Y.T.; Landesman, Y.; Acharya, C.; Calle, Y.; Zhong, M.Y.; Cea, M.; Tannenbaum, D.; Cagnetta, A.; Reagan, M.; Munshi, A.A.; et al. CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: Molecular mechanisms and therapeutic implications. Leukemia 2014, 28, 155–165. [Google Scholar] [CrossRef]
  258. Bahlis, N.J.; Sutherland, H.; White, D.; Sebag, M.; Lentzsch, S.; Kotb, R.; Venner, C.P.; Gasparetto, C.; Del Col, A.; Neri, P.; et al. Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood 2018, 132, 2546–2554. [Google Scholar] [CrossRef]
  259. Choudhary, C.; Kumar, C.; Gnad, F.; Nielsen, M.L.; Rehman, M.; Walther, T.C.; Olsen, J.V.; Mann, M. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 2009, 325, 834–840. [Google Scholar] [CrossRef]
  260. Weichert, W. HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 2009, 280, 168–176. [Google Scholar] [CrossRef]
  261. Tandon, N.; Ramakrishnan, V.; Kumar, S.K. Clinical use and applications of histone deacetylase inhibitors in multiple myeloma. Clin. Pharmacol. 2016, 8, 35–44. [Google Scholar] [CrossRef] [PubMed]
  262. Imai, Y.; Maru, Y.; Tanaka, J. Action mechanisms of histone deacetylase inhibitors in the treatment of hematological malignancies. Cancer Sci. 2016, 107, 1543–1549. [Google Scholar] [CrossRef] [PubMed]
  263. Bose, P.; Gandhi, V.; Konopleva, M. Pathways and mechanisms of venetoclax resistance. Leuk Lymphoma 2017, 58, 2026–2039. [Google Scholar] [CrossRef] [PubMed]
  264. Willis, S.N.; Fletcher, J.I.; Kaufmann, T.; van Delft, M.F.; Chen, L.; Czabotar, P.E.; Ierino, H.; Lee, E.F.; Fairlie, W.D.; Bouillet, P.; et al. Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak. Science 2007, 315, 856–859. [Google Scholar] [CrossRef]
  265. Kuwana, T.; Bouchier-Hayes, L.; Chipuk, J.E.; Bonzon, C.; Sullivan, B.A.; Green, D.R.; Newmeyer, D.D. BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Mol. Cell 2005, 17, 525–535. [Google Scholar] [CrossRef]
  266. Paner, A.; Patel, P.; Dhakal, B. The evolving role of translocation t(11;14) in the biology, prognosis, and management of multiple myeloma. Blood Rev. 2020, 41, 100643. [Google Scholar] [CrossRef]
  267. Kaufman, J.L.; Gasparetto, C.; Schjesvold, F.H.; Moreau, P.; Touzeau, C.; Facon, T.; Boise, L.H.; Jiang, Y.; Yang, X.; Dunbar, F.; et al. Targeting BCL-2 with venetoclax and dexamethasone in patients with relapsed/refractory t(11;14) multiple myeloma. Am. J. Hematol. 2021, 96, 418–427. [Google Scholar] [CrossRef]
  268. Lasica, M.; Anderson, M.A. Review of Venetoclax in CLL, AML and Multiple Myeloma. J. Pers. Med. 2021, 11, 463. [Google Scholar] [CrossRef]
  269. Khouri, J.; Faiman, B.M.; Grabowski, D.; Mahfouz, R.Z.; Khan, S.N.; Wei, W.; Valent, J.; Dean, R.; Samaras, C.; Jha, B.K.; et al. DNA methylation inhibition in myeloma: Experience from a phase 1b study of low-dose continuous azacitidine in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. Semin. Hematol. 2021, 58, 45–55. [Google Scholar] [CrossRef]
  270. Lai, A.C.; Crews, C.M. Induced protein degradation: An emerging drug discovery paradigm. Nat. Rev. Drug Discov. 2017, 16, 101–114. [Google Scholar] [CrossRef]
  271. An, S.; Fu, L. Small-molecule PROTACs: An emerging and promising approach for the development of targeted therapy drugs. EBioMedicine 2018, 36, 553–562. [Google Scholar] [CrossRef] [PubMed]
  272. Filippakopoulos, P.; Qi, J.; Picaud, S.; Shen, Y.; Smith, W.B.; Fedorov, O.; Morse, E.M.; Keates, T.; Hickman, T.T.; Felletar, I.; et al. Selective inhibition of BET bromodomains. Nature 2010, 468, 1067–1073. [Google Scholar] [CrossRef] [PubMed]
  273. Winter, G.E.; Buckley, D.L.; Paulk, J.; Roberts, J.M.; Souza, A.; Dhe-Paganon, S.; Bradner, J.E. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015, 348, 1376–1381. [Google Scholar] [CrossRef] [PubMed]
  274. Zhang, X.; Lee, H.C.; Shirazi, F.; Baladandayuthapani, V.; Lin, H.; Kuiatse, I.; Wang, H.; Jones, R.J.; Berkova, Z.; Singh, R.K.; et al. Protein targeting chimeric molecules specific for bromodomain and extra-terminal motif family proteins are active against pre-clinical models of multiple myeloma. Leukemia 2018, 32, 2224–2239. [Google Scholar] [CrossRef]
  275. Lim, S.L.; Damnernsawad, A.; Shyamsunder, P.; Chng, W.J.; Han, B.C.; Xu, L.; Pan, J.; Pravin, D.P.; Alkan, S.; Tyner, J.W.; et al. Proteolysis targeting chimeric molecules as therapy for multiple myeloma: Efficacy, biomarker and drug combinations. Haematologica 2019, 104, 1209–1220. [Google Scholar] [CrossRef]
  276. Lu, X.; Sabbasani, V.R.; Osei-Amponsa, V.; Evans, C.N.; King, J.C.; Tarasov, S.G.; Dyba, M.; Das, S.; Chan, K.C.; Schwieters, C.D.; et al. Structure-guided bifunctional molecules hit a DEUBAD-lacking hRpn13 species upregulated in multiple myeloma. Nat. Commun. 2021, 12, 7318. [Google Scholar] [CrossRef]
  277. Wang, M.; Kaufman, R.J. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat. Rev. Cancer 2014, 14, 581–597. [Google Scholar] [CrossRef]
  278. Fonseca, R.; Bergsagel, P.L.; Drach, J.; Shaughnessy, J.; Gutierrez, N.; Stewart, A.K.; Morgan, G.; Van Ness, B.; Chesi, M.; Minvielle, S.; et al. International Myeloma Working Group molecular classification of multiple myeloma: Spotlight review. Leukemia 2009, 23, 2210–2221. [Google Scholar] [CrossRef]
  279. Freedman, R.B.; Hirst, T.R.; Tuite, M.F. Protein disulphide isomerase: Building bridges in protein folding. Trends Biochem. Sci. 1994, 19, 331–336. [Google Scholar] [CrossRef]
  280. Laurindo, F.R.; Pescatore, L.A.; Fernandes Dde, C. Protein disulfide isomerase in redox cell signaling and homeostasis. Free Radic. Biol. Med. 2012, 52, 1954–1969. [Google Scholar] [CrossRef]
  281. Turano, C.; Coppari, S.; Altieri, F.; Ferraro, A. Proteins of the PDI family: Unpredicted non-ER locations and functions. J. Cell. Physiol. 2002, 193, 154–163. [Google Scholar] [CrossRef] [PubMed]
  282. Beer, D.G.; Kardia, S.L.; Huang, C.C.; Giordano, T.J.; Levin, A.M.; Misek, D.E.; Lin, L.; Chen, G.; Gharib, T.G.; Thomas, D.G.; et al. Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat. Med. 2002, 8, 816–824. [Google Scholar] [CrossRef] [PubMed]
  283. Nam, S.M.; Jeon, Y.J. Proteostasis In The Endoplasmic Reticulum: Road to Cure. Cancers 2019, 11, 1739. [Google Scholar] [CrossRef]
  284. Hetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102. [Google Scholar] [CrossRef] [PubMed]
  285. Hideshima, T.; Richardson, P.; Chauhan, D.; Palombella, V.J.; Elliott, P.J.; Adams, J.; Anderson, K.C. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001, 61, 3071–3076. [Google Scholar]
  286. Hasipek, M.; Grabowski, D.; Guan, Y.; Alugubelli, R.R.; Tiwari, A.D.; Gu, X.; DeAvila, G.A.; Silva, A.S.; Meads, M.B.; Parker, Y.; et al. Therapeutic Targeting of Protein Disulfide Isomerase PDIA1 in Multiple Myeloma. Cancers 2021, 13, 2649. [Google Scholar] [CrossRef]
  287. Vatolin, S.; Phillips, J.G.; Jha, B.K.; Govindgari, S.; Hu, J.; Grabowski, D.; Parker, Y.; Lindner, D.J.; Zhong, F.; Distelhorst, C.W.; et al. Novel Protein Disulfide Isomerase Inhibitor with Anticancer Activity in Multiple Myeloma. Cancer Res. 2016, 76, 3340–3350. [Google Scholar] [CrossRef]
  288. Robinson, R.M.; Reyes, L.; Duncan, R.M.; Bian, H.; Reitz, A.B.; Manevich, Y.; McClure, J.J.; Champion, M.M.; Chou, C.J.; Sharik, M.E.; et al. Inhibitors of the protein disulfide isomerase family for the treatment of multiple myeloma. Leukemia 2019, 33, 1011–1022. [Google Scholar] [CrossRef]
  289. Cohen, Y.C.; Zada, M.; Wang, S.Y.; Bornstein, C.; David, E.; Moshe, A.; Li, B.; Shlomi-Loubaton, S.; Gatt, M.E.; Gur, C.; et al. Identification of resistance pathways and therapeutic targets in relapsed multiple myeloma patients through single-cell sequencing. Nat. Med. 2021, 27, 491–503. [Google Scholar] [CrossRef]
  290. Lang, S.; Pfeffer, S.; Lee, P.H.; Cavalié, A.; Helms, V.; Förster, F.; Zimmermann, R. An Update on Sec61 Channel Functions, Mechanisms, and Related Diseases. Front. Physiol. 2017, 8, 887. [Google Scholar] [CrossRef]
  291. Domenger, A.; Choisy, C.; Baron, L.; Mayau, V.; Perthame, E.; Deriano, L.; Arnulf, B.; Bories, J.C.; Dadaglio, G.; Demangel, C. The Sec61 translocon is a therapeutic vulnerability in multiple myeloma. EMBO Mol. Med. 2022, 14, e14740. [Google Scholar] [CrossRef] [PubMed]
  292. Ng, Y.L.D.; Ramberger, E.; Bohl, S.R.; Dolnik, A.; Steinebach, C.; Conrad, T.; Müller, S.; Popp, O.; Kull, M.; Haji, M.; et al. Proteomic profiling reveals CDK6 upregulation as a targetable resistance mechanism for lenalidomide in multiple myeloma. Nat. Commun. 2022, 13, 1009. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Five-Year Relative Survival Percent, as reported at the Surveillance, Epidemiology, and End Results (SEER) Program(SEER Cancer Statistics Review, 1975–2018, National Cancer Institute. Bethesda, MD, USA, https://seer.cancer.gov/csr/1975_2018/ (accessed on 22 July 2022), based on November 2020 SEER data submission, posted to the SEER web site). The overall survival of MM dramatically improved after the development of novel proteasome inhibitors, immunomodulators and monoclonal antibodies. However, the survival advantage has plateaued over the past several years, emphasizing the need for discovery of new therapeutic modalities.
Figure 1. Five-Year Relative Survival Percent, as reported at the Surveillance, Epidemiology, and End Results (SEER) Program(SEER Cancer Statistics Review, 1975–2018, National Cancer Institute. Bethesda, MD, USA, https://seer.cancer.gov/csr/1975_2018/ (accessed on 22 July 2022), based on November 2020 SEER data submission, posted to the SEER web site). The overall survival of MM dramatically improved after the development of novel proteasome inhibitors, immunomodulators and monoclonal antibodies. However, the survival advantage has plateaued over the past several years, emphasizing the need for discovery of new therapeutic modalities.
Cancers 14 04082 g001
Figure 2. Clinical definitions, risk stratification, and available therapies for multiple myeloma and preexisting plasma cell disorders. End organ damage is defined as: Hypercalcemia (serum calcium > 1 mg/dL higher than the upper limit of normal or >11 mg/dL); renal insufficiency, creatinine clearance < 40 mL/minute or serum creatinine > 2 mg/dL; anemia with a hemoglobin value > 2 g/dL below the lowest limit of normal or a hemoglobin value < 10.0 g/dL; and Bone lesions, ≥1 osteolytic lesion on imaging. Abbreviations: M, monoclonal; BMPC, Bone Marrow Plasma Cells; FLC, Free Light Chain; B2M, Beta-2 Microglobulin; CA, Cytogenetics, IMiD, Immunomodulators; AHCT, Autologous Hematopoietic Cell Transplantation; CAR, Chimeric Antigen Receptor.
Figure 2. Clinical definitions, risk stratification, and available therapies for multiple myeloma and preexisting plasma cell disorders. End organ damage is defined as: Hypercalcemia (serum calcium > 1 mg/dL higher than the upper limit of normal or >11 mg/dL); renal insufficiency, creatinine clearance < 40 mL/minute or serum creatinine > 2 mg/dL; anemia with a hemoglobin value > 2 g/dL below the lowest limit of normal or a hemoglobin value < 10.0 g/dL; and Bone lesions, ≥1 osteolytic lesion on imaging. Abbreviations: M, monoclonal; BMPC, Bone Marrow Plasma Cells; FLC, Free Light Chain; B2M, Beta-2 Microglobulin; CA, Cytogenetics, IMiD, Immunomodulators; AHCT, Autologous Hematopoietic Cell Transplantation; CAR, Chimeric Antigen Receptor.
Cancers 14 04082 g002
Figure 3. The basic principles of the immunotherapies in MM and their place in the current treatment landscape. CAR T cells are T cells genetically modified with the use of a viral vector to express a chimeric antigen receptor on their surface, which targets specific tumor antigens of malignant plasma cells. Similarly, bispecific antibodies are monoclonal antibodies targeting both an antigen on the malignant MM cells and simultaneously an antigen on the surface of physiologic T cells, creating an immunologic bridge. ADC are monoclonal antibodies against antigenic epitopes on the surface on MM cells, carrying a cytotoxic payload. The binding of the above agents to their antigenic targets on malignant MM cells leads to activation of the immune system, with subsequent destruction of the MM cells.
Figure 3. The basic principles of the immunotherapies in MM and their place in the current treatment landscape. CAR T cells are T cells genetically modified with the use of a viral vector to express a chimeric antigen receptor on their surface, which targets specific tumor antigens of malignant plasma cells. Similarly, bispecific antibodies are monoclonal antibodies targeting both an antigen on the malignant MM cells and simultaneously an antigen on the surface of physiologic T cells, creating an immunologic bridge. ADC are monoclonal antibodies against antigenic epitopes on the surface on MM cells, carrying a cytotoxic payload. The binding of the above agents to their antigenic targets on malignant MM cells leads to activation of the immune system, with subsequent destruction of the MM cells.
Cancers 14 04082 g003
Figure 4. Underlying mechanism of action of selected targeted therapies used for MM in the clinical setting. Venetoclax works by primarily binding to the BCL-2 anti-apoptotic protein, allowing the activation of BAK and subsequently caspases leading to MM apoptosis. Selinexor blocks the transport of vital proteins and other molecules from the nucleus to the cytoplasm of the MM cells, leading to cell death. Histone deacetylation inhibitors act at an epigenetic level, blocking the deacetylation of the DNA in the nucleus of the malignant cell.
Figure 4. Underlying mechanism of action of selected targeted therapies used for MM in the clinical setting. Venetoclax works by primarily binding to the BCL-2 anti-apoptotic protein, allowing the activation of BAK and subsequently caspases leading to MM apoptosis. Selinexor blocks the transport of vital proteins and other molecules from the nucleus to the cytoplasm of the MM cells, leading to cell death. Histone deacetylation inhibitors act at an epigenetic level, blocking the deacetylation of the DNA in the nucleus of the malignant cell.
Cancers 14 04082 g004
Figure 5. Heightened PDIA1 is critical to maintain ER homeostasis in MM Cells. Disruption of the ER causes dysregulation of the protein folding process, known as ER stress, secondary to the accumulation of unfolded or misfolded proteins. This leads to activation of the unfolded protein response (UPR), with subsequent activation of downstream pathways inducing apoptosis. Abbreviations: PDIA1, protein disulfide isomerase inhibitor A1; ER, endoplasmic reticulum; UPR, unfolded protein response.
Figure 5. Heightened PDIA1 is critical to maintain ER homeostasis in MM Cells. Disruption of the ER causes dysregulation of the protein folding process, known as ER stress, secondary to the accumulation of unfolded or misfolded proteins. This leads to activation of the unfolded protein response (UPR), with subsequent activation of downstream pathways inducing apoptosis. Abbreviations: PDIA1, protein disulfide isomerase inhibitor A1; ER, endoplasmic reticulum; UPR, unfolded protein response.
Cancers 14 04082 g005
Table 1. Major clinical trials of the standard-of-care regimens for MM currently used in routine clinical practice.
Table 1. Major clinical trials of the standard-of-care regimens for MM currently used in routine clinical practice.
RegimenTrial Name NCT NumberPhaseNDisease StatusOutcomes
Dara-Vd vs. VdCASTOR
NCT02136134
3498RRMMORR: 83.8% vs. 63.2% (p < 0.0001)
≥CR: 28.8% vs. 9.8% (p < 0.0001)
m-PFS: 16.7 vs. 7.1 months (p < 0.0001)
Dara-Rd
vs. Rd
POLLUX
NCT02076009
3569RRMMORR: 92.9 vs. 76.4% (p < 0.0001)
≥CR: 56.6 vs. 23.2% (p < 0.0001
m-PFS: 44.5 vs. 17.5 months (p < 0.0001)
Dara-Kd
vs. Kd
CANDOR
NCT03158688
3446RRMMORR: 84% vs. 75% (p = 0.0080)
≥CR: 33% vs. 13%
m-PFS: 28.6 vs. 15.2 months (p < 0.0001)
Dara-Pd
vs. Pd
APOLLO
NCT03180736
3304RRMMORR: 69% vs. 46% (p < 0.0001)
≥CR: 25 vs. 4%, (p < 0.0001)
m-PFS: 12.4 vs. 6.9 months (p = 0.0018)
Dara-Rd
vs. RD
MAIA
NCT02252172
3737NDMMORR: 92.9% vs. 81.6 (p < 0.0001)
≥CR: 51% vs. 30% (p < 0.0001)
m-PFS: NR vs. 34.4 months (p < 0.0001)
Dara-VMP vs. VMPALCYONE
(NCT02195479)
3706NDMMORR: 90.9% vs. 73.9% (p < 0.0001)
≥CR: 46% vs. 25% (p < 0.0001)
m-PFS: 36.4 vs. 19.3 months (p < 0.0001)
Dara-VTd
vs. VTd
CASSIOPEIA
NCT02541383
31085NDMMAt day 100 post AHCT:
CR: 29% vs. 20% (p = 0.001)
≥CR: 39% vs. 26% (p < 0.0001)
m-PFS: NR for both groups (p < 0.0001)
Dara-VRd
vs. VRd
GRIFFIN
NCT03710603
2207
NDMMAt the end of post-AHCT consolidation:
ORR: 99% vs. 91.8 (p = 0.016)
sCR: 42.4% vs. 32% (p = 0.068)
m-PFS: NR for both groups
Isa-Pd vs. PdICARIA
NCT02990338
3307RRMMORR: 60% vs. 35% (p < 0.0001)
m-PFS: 11.5 vs. 6.5 months (p = 0.001)
m-OS: 24.6 vs. 17.7 months (p = 0.028)
Isa-Kd vs. KdIKEMA
NCT03275285
3302RRMMCR: 40% vs. 28%
≥VGPR: 73% vs. 56% (p = 0.0011)
m-PFS: 19.15 months vs. NR (p = 0.0007)
Elo-Rd
vs. Rd
ELOQUENT-2
NCT01239797
3321
RRMMORR: 79%, vs. 66% (p < 0.001)
m-PFS: 19.4 vs. 14.9 months (p < 0.001)
m-OS: 48.3 vs. 39.6 months (p =  0.0408)
Elo-Pd
vs. Pd
ELOQUENT-3
NCT02654132
2117RRMMORR: 53% vs. 26%
m-PFS: 10.3 vs. 4.7 months (p = 0.008)
m-OS: 29.8 vs. 17.4 months (p = 0.0217)
Abbreviations: dara, daratumumab; Vd, bortezomib–dexamethasone; Rd, lenalidomide–dexamethasone; Kd, carfilzomib–dexamethasone; Pd, pomalidomide–dexamethasone; VMP, bortezomib–melphalan–dexamethasone; VTd, bortezomib–thalidomide–dexamethasone; VRd, bortezomib–lenalidomide–dexamethasone; Isa, isatuximab; m-PFS, median progression-free survival; m-OS, median overall survival.
Table 2. Major CAR T cell and Bispecific Antibody Clinical Trials with reported outcomes in MM.
Table 2. Major CAR T cell and Bispecific Antibody Clinical Trials with reported outcomes in MM.
Agent NameTarget
Antigen
NCT Number (Trial Name)PhaseNDisease Status OutcomesCRS ICANS
CAR -T Cell Therapy Products
bb21217BCMANCT03274219
(CRB-402)
146RRMM▪ ORR: 55%, ≥CR: 18%, VGPR: 30%
▪ mPFS: 7.2 months
▪ CRS: 67%
▪ ICANS: 22%
Idecabtagene vicleucelBCMANCT02658929
(CRB-401)
162RRMM▪ ORR: 76%, ≥CR: 39%, ≥VGPR: 65%
▪ mPFS/mOS: 8.8/34.2 months
▪ CRS: 76%
▪ ICANS: 44%
NCT03361748
(KarMMa)
2128RRMM▪ ORR: 73%, ≥CR: 33%, ≥VGPR: 52%
▪ mPFS/mOS: 8.8/19.4 months
▪ CRS: 84%
▪ ICANS: 18%
Ciltacabtagene autoleucelBCMANCT03548207 (CARTITUDE-1)1/2113RRMM▪ ORR: 98%, CR: 82.5%, VGPR: 95%
▪ mPFS and mOS: NR
▪ CRS: 95%
▪ ICANS: 21%
NCT04133636 (CARTITUDE-2)220RRMM▪ ORR: 95%, CR: 75%, ≥VGPR: 85%
▪ mPFS and mOS: NR
▪ CRS: 85%
▪ ICANS: 20%
Zevorcabtagene autoleucelBCMANCT03716856
NCT03302403
NCT03380039
124RRMM▪ ORR: 87.5%, ≥CR: 80%
▪ mPFS 9.2 months
▪ CRS: 62.5%
▪ ICANS: 12.5%
NCT03975907
(LUMMICAR-1)
1/238RRMM▪ ORR: 92%, CR: 79%
▪ mPFS: 22.7 months
▪ CRS: 73.7%
NCT03915184
(LUMMICAR-2)
1/234RRMM▪ ORR: 100%
▪ mPFS and mOS: NR
▪ CRS: 86%
Orvacabtagene autoleucel (JCARH125)BCMANCT03090659
(LEGEND-2)
174RRMM▪ ORR 88%, CR 73%
▪ mPFS: 18 months, mOS: NR
▪ CRS: 92%
▪ ICANS: 21%
NCT03430011
(EVOLVE)
1/2115RRMM▪ ORR: 82%
▪ mPFS: NR
▪ CRS: 75%
CT103ABCMANCT05066646
(FUMANBA-1)
1/279RRMM▪ ORR: 94.9%
▪ CR/sCR: 69.6%
▪ CRS 94.9%
▪ ICANS: 2.5%
CART-ddBCMABCMANCT04155749125RRMM▪ ORR: 100%, ≥CR: 75%
▪ mPFS and mOS: NR
▪ CRS: 100%
▪ ICANS: 16%
GC012FBCMA/CD19NCT04236011
NCT04182581
128RRMM▪ ORR: 80–100%▪ CRS: 100%
OriCAR-017GPRC5DNCT05016778111RRMM▪ ORR: 100%
▪ MRD (10−5) negativity: 100%
▪ CRS: 100%
Bispecific Antibodies
TeclistamabBCMAxCD3NCT03145181 NCT04557098
(MajesTEC-1)
1/2165RRMM▪ ORR: 63%, ≥CR 39.4%
▪ MRD (10−5) negativity: 26.7%
▪ mPFS: 11.3 months
▪ CRS: 72%
▪ ICANS: 3%
Teclistamab + daratumumabNCT04108195
(TRIMM-2)
1b46RRMM▪ ORR: 78%
▪ ≥VGPR: 73%
▪ CRS: 61%
▪ ICANS: 2.1%
ElranatamabBCMAxCD3NCT03269136
(MagnetisMM-1)
155RRMM▪ ORR: 64%▪ CRS: 67%
NCT04649359
(MagnetisMM-3)
260RRMM▪ ORR: NR▪ CRS 58.9%
▪ ICANS: 3.6%
TalquetamabGPRC5DxCD3NCT03399799
(MonumenTAL-1)
1174RRMM▪ ORR 63%
▪ ≥VGPR 50%
▪ CRS 79%
▪ ICANS: 7%
Abbreviations: ORR, overall response rate; CR, complete response; VGPR, very good partial response rate; mPFS, median progression-free survival; mOS, median overall survival; RRMM, relapsed/refractory multiple myeloma; CRS, cytokine release syndrome; NR, not reached.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dima, D.; Jiang, D.; Singh, D.J.; Hasipek, M.; Shah, H.S.; Ullah, F.; Khouri, J.; Maciejewski, J.P.; Jha, B.K. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers 2022, 14, 4082. https://doi.org/10.3390/cancers14174082

AMA Style

Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers. 2022; 14(17):4082. https://doi.org/10.3390/cancers14174082

Chicago/Turabian Style

Dima, Danai, Dongxu Jiang, Divya Jyoti Singh, Metis Hasipek, Haikoo S. Shah, Fauzia Ullah, Jack Khouri, Jaroslaw P. Maciejewski, and Babal K. Jha. 2022. "Multiple Myeloma Therapy: Emerging Trends and Challenges" Cancers 14, no. 17: 4082. https://doi.org/10.3390/cancers14174082

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