You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Multiple Myeloma Therapy
Edit

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. 

multiple myeloma immunotherapy targeted therapy

1. Immunotherapy

1.1. 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 [1][2]. 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 [3][4][5][6][7][8][9][10]. 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 [11][12].
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 [13][14][15][16][17][18]. 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 [14][18][19][20][21]. 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.

1.2. 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.

1.3. 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 [22][23]. 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 [24][25]. 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 [23][24][26]. Experiments have shown that PD-L1, the major ligand of the PD-1 receptor, can be upregulated on malignant PCs [26]. MM cells with high PD-L1 expression appear to be more proliferative and resistant to therapy, indicating increased aggressiveness [27]. 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 [28][29][30][31]. 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 [32].
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 [32][33]; 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 [34]. 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 [35].
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 [36][37][38]. Anti-PD-L1 inhibitors have also been used in combination with daratumumab without safety warnings but no clinical benefit so far [39][40][41][42]. 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.

1.4. 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 [43][44]. 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 [44] (Figure 1). 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 [44]. 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 [45]. ADC can also exert their effects via ADCC, ADCP, or CDC [46][47].
Figure 1. 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.

1.5. 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 [48] (Figure 1). 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 [49]. GPRC5D is a G-protein–coupled receptor with unclear function that is highly expressed on the surface of myeloma cells [50]. FcRH5 belongs to the immunoglobulin superfamily and is located only on the surface of B cells with increasing expression on myeloma cells [51][52]. At present, all bsAbs in clinical trials target the CD3 on the surface of T cells [49]. However, in a preclinical level, bispecific NK-cell engagers are also under investigation with good anti-myeloma activity [53][54][55].

1.6. 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 [56][57]. 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 [58][59][60] (Figure 1). First-generation CAR lacked a costimulatory domain, resulting in only moderate responses [61]. 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 [62].
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 [63][64]. The CAR gene is subsequently transcribed and expressed as a surface receptor [65][66]. CAR T cell manufacturing occurs ex vivo and takes 4 weeks on average [65][66]. 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 [67][68].
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 [65]. BCMA was the first antigen to be targeted in CAR T cell therapy clinical trials [69]. 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 [70][71]. 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 [72][73]. 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 [74][75].
Despite the associated high responses, not all patients have durable responses after CAR T cell therapy [76], which is related to several tumor- and CAR-T-cell-construct-related factors [76]. 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 [77]. 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 [78][79]. One mechanism that could explain antigenic loss is acquired biallelic BCMA deletion, resulting in decreased BCMA expression [80][81]. High tumor load also appears to negatively affect the efficacy of CAR T cell therapy, perhaps due to CAR T cell exhaustion [70][82]. High expression of immune checkpoints on the surface on myeloma and CAR T cells can also attenuate CAR T cell activity [60][83][84]. 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 [85][86].
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 [87][88][89] and a high CD4/CD8 T cell ratio in the apheresis collection [68][89], which is typically seen in patients early in their disease course [90], leads to higher CAR T cell proliferation, expansion, and persistence and subsequently higher response rates [91][92]. 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 [93]. Several preclinical studies are currently investigating simultaneous targeting of BCMA and SLAMF7, GPRC5D, or CD38, molecules uniformly expressed on MM cells [85][93][94][95]. 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 [96][97][98]. 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. [82][99][100]. 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 [101].

1.7. 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. [102]. 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 [103][104][105]. 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. [103][104][105]. 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) [102]. 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 [106][107]. 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. [107]. 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 [108]. 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 [109]. As previously mentioned, upregulation of PD-1/PD-L1 [25][110][111] is associated with poor prognosis in patients with MM [112]. Stimulation with the IO103 peptide stimulated PD-L1-specific T cytotoxic cells against PD-L1-expressing MM cells. In this research, 10 patients with MM who were 6 months post AHCT were enrolled [109]. 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) [109].
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 [113]. 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 [114]. 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).

2. Targeted Therapies and Small Molecules

2.1. Exportin Inhibitors

The nuclear pore complexes (NPC) are large cylindrical channels, composed of several copies of >30 different proteins called nucleoporins [115]. 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. [116]. 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 [117][118]. 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 [119]. 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 [120][121].
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. [106] 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 [122][123][124] (Figure 2). It has also been shown to block NF-kB, which regulates osteoclast differentiation [125]. 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 [126].
Figure 2. 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.

2.2. 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 [127] (Figure 2). In MM, overexpression of HDAC, especially HDAC-1, has been associated with a poor prognosis and with resistance to PI [128][129]. 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 [130].
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.

2.3. 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 [131]. The latter subcategory primarily works by binding to anti-apoptotic proteins, activating the BAX/BAK proteins, directly or indirectly, and inducing apoptosis [132][133]. 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 [134]. 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 2) 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 [135]. As a result, translocation (11;14) has emerged as the first predictor of susceptibility to Bcl-2 inhibition in MM patients [136]. Venetoclax is not FDA-approved yet; however, the NCCN guidelines recommend its use in RRMM with the translocation (11;14).

2.4. 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 [137]. 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).

2.5. 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) [138]. 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) [138][139].
An initial PROTAC called dBET1 was constructed using thalidomide (as an E3 ligase binding domain) and JQ1 (as a POI binding domain) [140]. 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 [141]. 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 [142]. 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 [143]. Another, newer experimental PROTAC targeting the proteasome substrate receptor hRpn13, which was found to be upregulated in MM, was tested in in vitro studies [144]. Optimization of PROTAC design for potential clinical development is eagerly awaited.

References

  1. 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.
  2. 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.
  3. Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692.
  4. Vyas, M.; Müller, R.; Pogge von Strandmann, E. Antigen Loss Variants: Catching Hold of Escaping Foes. Front. Immunol. 2017, 8, 175.
  5. 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.
  6. Dhatchinamoorthy, K.; Colbert, J.D.; Rock, K.L. Cancer Immune Evasion Through Loss of MHC Class I Antigen Presentation. Front. Immunol. 2021, 12, 636568.
  7. 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.
  8. 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.
  9. Kumar, S.; Kimlinger, T.; Morice, W. Immunophenotyping in multiple myeloma and related plasma cell disorders. Best Pract. Res. Clin. Haematol. 2010, 23, 433–451.
  10. 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.
  11. 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.
  12. 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.
  13. 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.
  14. 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.
  15. 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.
  16. 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.
  17. 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.
  18. 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.
  19. 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.
  20. 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.
  21. 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.
  22. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264.
  23. Zou, W.; Chen, L. Inhibitory B7-family molecules in the tumour microenvironment. Nat. Rev. Immunol. 2008, 8, 467–477.
  24. 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.
  25. 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.
  26. 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- and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007, 110, 296–304.
  27. 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.
  28. 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.
  29. 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.
  30. 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.
  31. Asimakopoulos, F. TIGIT checkpoint inhibition for myeloma. Blood 2018, 132, 1629–1630.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. Tsuchikama, K.; An, Z. Antibody-drug conjugates: Recent advances in conjugation and linker chemistries. Protein Cell 2018, 9, 33–46.
  44. Yu, B.; Liu, D. Antibody-drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma. J. Hematol. Oncol. 2019, 12, 94.
  45. Herrera, A.F.; Molina, A. Investigational Antibody-Drug Conjugates for Treatment of B-lineage Malignancies. Clin. Lymphoma Myeloma Leuk 2018, 18, 452–468.e454.
  46. 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.
  47. 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.
  48. Kontermann, R.E.; Brinkmann, U. Bispecific antibodies. Drug Discov. Today 2015, 20, 838–847.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. Sadelain, M.; Brentjens, R.; Rivière, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013, 3, 388–398.
  57. 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.
  58. Sadelain, M.; Rivière, I.; Riddell, S. Therapeutic T cell engineering. Nature 2017, 545, 423–431.
  59. 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.
  60. 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.
  61. 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.
  62. Mikkilineni, L.; Kochenderfer, J.N. Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood 2017, 130, 2594–2602.
  63. Lim, W.A.; June, C.H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168, 724–740.
  64. 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.
  65. Srivastava, S.; Riddell, S.R. Engineering CAR-T cells: Design concepts. Trends Immunol. 2015, 36, 494–502.
  66. Wang, X.; Rivière, I. Clinical manufacturing of CAR T cells: Foundation of a promising therapy. Mol. Ther. Oncolytics 2016, 3, 16015.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. 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.
  95. 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.
  96. 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.
  97. 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.
  98. 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.
  99. 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.
  100. 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.
  101. 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.
  102. 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.
  103. 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.
  104. 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.
  105. 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.
  106. 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.
  107. 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.
  108. 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.
  109. 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.
  110. 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.
  111. 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.
  112. 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.
  113. 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.
  114. 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.
  115. 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.
  116. Theodoropoulos, N.; Lancman, G.; Chari, A. Targeting Nuclear Export Proteins in Multiple Myeloma Therapy. Target. Oncol. 2020, 15, 697–708.
  117. Azizian, N.G.; Li, Y. XPO1-dependent nuclear export as a target for cancer therapy. J. Hematol. Oncol. 2020, 13, 61.
  118. Turner, J.G.; Dawson, J.; Sullivan, D.M. Nuclear export of proteins and drug resistance in cancer. Biochem. Pharmacol. 2012, 83, 1021–1032.
  119. 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.
  120. 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.
  121. 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.
  122. 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.
  123. 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.
  124. Vogt, P.K.; Jiang, H.; Aoki, M. Triple layer control: Phosphorylation, acetylation and ubiquitination of FOXO proteins. Cell Cycle 2005, 4, 908–913.
  125. 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.
  126. 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.
  127. 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.
  128. Weichert, W. HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 2009, 280, 168–176.
  129. Tandon, N.; Ramakrishnan, V.; Kumar, S.K. Clinical use and applications of histone deacetylase inhibitors in multiple myeloma. Clin. Pharmacol. 2016, 8, 35–44.
  130. Imai, Y.; Maru, Y.; Tanaka, J. Action mechanisms of histone deacetylase inhibitors in the treatment of hematological malignancies. Cancer Sci. 2016, 107, 1543–1549.
  131. Bose, P.; Gandhi, V.; Konopleva, M. Pathways and mechanisms of venetoclax resistance. Leuk Lymphoma 2017, 58, 2026–2039.
  132. 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.
  133. 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.
  134. 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.
  135. 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.
  136. Lasica, M.; Anderson, M.A. Review of Venetoclax in CLL, AML and Multiple Myeloma. J. Pers. Med. 2021, 11, 463.
  137. 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.
  138. Lai, A.C.; Crews, C.M. Induced protein degradation: An emerging drug discovery paradigm. Nat. Rev. Drug Discov. 2017, 16, 101–114.
  139. An, S.; Fu, L. Small-molecule PROTACs: An emerging and promising approach for the development of targeted therapy drugs. EBioMedicine 2018, 36, 553–562.
  140. 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.
  141. 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.
  142. 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.
  143. 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.
  144. 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.
More
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , ,
View Times: 810
Revisions: 2 times (View History)
Update Date: 09 Sep 2022
Video Production Service