1. INTRODUCTION
Chimeric antigen receptor T (CAR-T) cell therapy exhibit impressive response rates in relapsed and refractory multiple myeloma (MM) patients, which led to FDA approval of two B cell maturation antigen (BCMA)-specific CAR-T cell therapies (ide-cel and cilta-cel) [1–4]. Other candidate CAR-T cell targets for MM include signaling lymphocytic activation molecule F7 (SLAMF7) [5], CD38 [6], CD138, CD19 [7], Lewis-Y, CD56, CD44v6, Kappa-light chain, and G-protein-coupled receptor class C group 5 member D (GPRC5D) [8]. The availability of these CAR-T cell products offers the hope of disease control and remission for patients lacking therapeutic options; however, the durable response has only been observed in a small group of patients [9]. Multi-functional TRUCK T cells (T-cells redirected for antigen unrestricted cytokine-initiated killing) or ‘the fourth-generation’ CAR T cells are now being evaluated in preclinical settings [10, 11]. The fourth-generation CAR-T cells incorporate cytokine-secreting vectors, such as IL-12, IL-18, or transcription factors-producing vectors, to modulate the tumor microenvironment (TME) and enhance the activation of CAR-T cells. Advances in genome editing techniques, such as CRISPR/CAS9, allows any array of genes in the genome to be incorporated or knocked out, which results in more individualized, efficient, and less toxic CAR-T cell products [12–14].
By combining the ability to recognize tumor-specific antigens in a non-major histocompatibility complex (MHC)-restricted manner with potent activation of T cells to kill the tumor cells, CAR-T cell therapy is considered one of the most promising choices for achieving a cure in MM patients; however, the efficacy of CAR-T cell therapy for hematologic malignancies is largely influenced by the TME [10, 15, 16]. The MM TME contains immunosuppressive cells, an extracellular matrix (ECM), and vasculature components that facilitate the development of tumors. The oxygen concentration and nutrient imbalance inhibit the activity of CAR-T cells. In this review, we summarized the challenges the TME poses to CAR-T cells, with a particular emphasis on MM intrinsic factors, including immunosuppressive cells, structural components, hypoxia, nutrient starvation, and metabolism. Moreover, we also discussed efforts underway to overcome these TME challenges in treating MM, taking into account the latest advances in the next-generation CAR-T cells and single-cell sequencing technology in recent years.
2. OVERCOMING IMMUNOSUPPRESSIVE CELLS
Impairment of immunosurveillance and immune effector function of the host immune system, including both adaptive and innate immunity, leads to immune escape in MM, and contributes to tumorigenesis and tumor progression [17]. Anti-tumor responses of the main effector immune cells, including natural killer (NK) cells [18] and cytotoxic T lymphocytes (CTLs) [19], are impaired in the TME. In addition, MM often induces an immunosuppressive TME, which promotes the expansion of myeloid-derived suppressor cells (MDSCs) [20], tumor-associated macrophages (TAMs) [21], and regulatory T cells (Tregs) [22]. Anti-inflammatory cytokines and chemokines secreted by these immunosuppressive cells, in turn, foster the TME and further promote immune escape and tumor growth [23, 24].
2.1 MDSCs
As a highly heterogeneous group of immature myeloid cells, MDSCs are capable of suppressing immune responses [25, 26]. There are two major subsets of MDSCs, as follows: monocytic-MDSCs (M-MDSCs), which are morphologically and phenotypically similar to monocytes; and polymorphonuclear or granulocytic-MDSCs (PMN-MDSCs), which resemble neutrophils. A subset of more immature progenitors (Lin-CD33+ cells) are a third group of MDSCs, and defined as “early-stage MDSCs (eMDSCs)” [27]. Previous studies have reported an increased number of MDSCs in MM patients compared to healthy donors [28]. In addition, an elevated level of peripheral blood (PB) monocytes correlates with poor prognosis in patients with solid tumors [29]. The humoral and cellular immunosuppressive effects of MDSCs have been elucidated in various hematologic malignancies, with T cells serving as the major targets [27]. MDSCs inhibit T cell activation by secreting reactive oxygen species (ROS) and nitric oxide (NO), and by scavenging amino acids, including arginine, cysteine, and tryptophan, which are essential for T cell proliferation [27, 28]. MDSCs also produce IL-10 and TGF-β to induce Tregs [15]. Moreover, the presence of MDSCs is correlated with patient outcome following treatment with with CAR-T cell therapy. In a phase I/IIa trial involving third-generation CAR-T cells, lymphoma and leukemia patients with lower levels of M-MDSCs had better responses than patients with higher levels of M-MDSCs [30].
Considering the adverse effect of MDSCs on CAR-T cell cytolytic function and proliferation, it is reasonable to designate MDSCs as a treatment target along with CAR-T cell therapy. Effective strategies can target MDSCs through MDSC depletion and deactivation, as well as blocking MDSC recruitment to the TME [25, 27]. As a proof of concept, it has been observed that complete depletion of MDSCs following anti-Gr-1 antibody treatment in EL-4 tumor-bearing mice results in improved anti-tumor efficacy of CAR-T cells [31]. A high amount of arginase-1 is expressed in activated MDSCs, thus revealing that the MDSC suppressive mechanisms are reversed by an inhibitor of arginase-1 in MM [28, 32]. Furthermore, expansion of MDSCs driven by granulocyte-macrophage colony-stimulating factor (GM-CSF) and STAT3 signaling, thus GM-CSF neutralization and STAT3 inhibition may be alternative targets for limiting the immunosuppressive activity of MDSCs [33]. Furthermore, accumulation of MDSCs is significantly reduced by targeting chemokines and cytokines, such as IL-17, exosomal CD47, and S100A8/S100A9, which have important roles in the trafficking of MDSCs to the TME [34]. To better understand the immune changes in the MM TME, Zavidij et al. [19] applied single-cell RNA sequencing (scRNA-seq) to profile the MM TME. Significant up-regulation of HLA-DR was noted for CD14+ monocytes in MM patients compared to healthy controls; however, the surface expression of CD14+ was significantly lower in MM patients. Such a phenotypic change in TME T CD14+ monocytes towards MDSCs is rescued after treatment with MARCHF1/MARCH1 knockdown.
2.2 TAMs
TAMs represent one of the main immune cells in the TME, the accumulation of which in the MM TME is associated with poor prognosis [35]. Based on the activation status, TAMs can be classified into M1 and M2 macrophages [36]. A newer terminology to describe TAMs has been proposed as “M1-like” and “M2-like” TAMs; the former is associated with pro-inflammatory role and “M2-like” TAMs have an anti-inflammatory and wound-healing role in the TME. “M1-like” TAMs are induced by type-1 cytokines, such as IFN-γ and TNF-α, while “M2-like” TAMs are activated by cytokines, including IL-13 and IL-4. The transcriptomic diversity of TAMs has recently been described by two large-scale cross-tissue scRNA-seq studies [37–39]. A phenotypic plasticity spectrum from pro- to anti-inflammatory TAMs has been demonstrated, and TAMs with angiogenic and proliferative functions are also preserved in almost all TMEs [37].
TAMs sustain tumor progression, tumor cell metastasis, and invasion in multiple ways. Other features of TAMs include secretion of growth factors, such as fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF), which promote proliferation of tumor cells, a variety of proteolytic enzymes (including metalloproteinases that facilitate matrix remodeling), and ROS and NO that induce genetic instability of hematologic malignancies [15]. TAMs also express immunosuppressive cytokines, including TGF-β and IL-10, that suppress T cell immune effects, leading to metabolic starvation of T cells by secreting amino acid-depleting enzymes, including 2,3-dioxygenase (IDO) and arginase-1 [40]. By expressing multiple immune checkpoint ligands, including programmed death-ligand 1 (PD-L1), PD-L2, and B7-H4, TAMs induce immunosuppression [40]. Zheng et al. [21] first showed that TAMs activate anti-apoptosis signaling of MM via physical interaction, thereby contributing to MM cell drug resistance.
The pro-tumorigenic characteristics of TAMs make TAMs potential targets when treating MM with CAR-T cell therapy. Emerging strategies to target TAMs include blockade of monocyte/macrophage recruitment to the bone marrow (BM), direct depletion of TAMs from the TME, and re-education of M2 TAMs into M1 TAMs [36, 37]. Several preclinical studies have shown antigen-specific phagocytosis by macrophages after modification with CAR (CAR-M) [41–43]. In a recent study Morrissey et al. [41] developed a series of CAR-Ms to phagocytose Raji B cells, CD19+, or CD22+ tumor cells. It was further shown that CAR-M combined with CD47 monoclonal antibody or SIRPα enhance the phagocytic function of CAR-M in vitro [41].
2.3 Tregs
Tregs are a subset of specialized CD4+ T cells that are capable of exerting strong immunosuppressive activity and expressing the transcription factor, FOXP3 [22]. Tregs have an important role in regulating the immune system and mediating immune tolerance for homeostasis [44, 45]; however, Tregs can be differentiated to suppress cytotoxicity of CD8+ T cells in the TME via the following mechanisms: secretion of immunosuppressive cytokines (TGF-β or IL-10); competitive consumption of IL-2; CTLA-4-mediated inhibition of antigen-presenting cells in the form of direct contact; and secretion of granzyme and/or perforin to lytic effector T cells [40].
Sufficient evidence suggests enrichment of Tregs in the PB of MM patients, and it has also been observed that the frequency of Tregs in PB is positively associated with disease risk stage and paraprotein level [46, 47]. Furthermore, an elevated Treg level is also an adverse prognostic factor for MM patients [46]. An obvious strategy to address the immunosuppressive limitation of Tregs is to specifically eliminate Tregs from the TME [45]. The number and function of Tregs have been shown to be reduced, accompanied by restoration of anti-tumor immunity after treatment with low-dose cyclophosphamide [48]. Tregs can be transiently removed from the TME and an improved survival rate can be achieved after treatment with low-dose cyclophosphamide in a murine model of MM [49]. In addition, emerging strategies targeting Tregs include targeting immune checkpoints on Tregs and reversing Tregs towards an anti-tumor immunity phenotype [45].
2.4 Other cellular compartments
Mesenchymal stem/stromal cells (MSCs) are a subset of multipotent stem cells that give rise to chondrocytes, adipocytes, osteocytes, and other lineages [50]. MSCs from BM are CD90-, CD73-, and CD105-positive, and negative for CD45, CD34, CD14, and HLA-DR [50, 51]. MSCs represent an integral component of the MM TME, and the presence of MSCs is essential for MM development, progression, and metastasis [52]. By using scRNA-seq, Jong et al. [53] identified two MSC clusters that are strongly enriched in MM BM, and differentially expressed genes of these MM-specific MSC clusters show significant enrichment for an inflammation-related pathway. A longitudinal analysis, however, has revealed that successful anti-tumor induction therapy does not revert BM inflammation, indicating the essential role of MSC-centric TME inflammation in MM persistence [53].
Osteoclasts are the critical mediator of bone absorption and are responsible for bone destruction in > 80% of MM patients [54, 55]. Osteoclasts in the MM TME are large multi-nucleated cells, which are differentiated from CD14+ myeloid cells and induced by OC-activating cytokines and receptor activators of nuclear factor-κB (RANKL) by BM supporting cells and MM cells [56]. Traditionally, osteoclasts are thought to be essential for regulation of bone metabolism [57]; however, accumulating evidence has shown that osteoclasts are important immunomodulatory compartments and are involved in the formation of an immunosuppressive MM TME [23, 56]. The expression of a variety of immunosuppressive molecules, including PD-L1, IDO, CD200, and Galectin-9, are upregulated during osteoclastogenesis. The proliferation of CD4+ and CD8+ T cells is significantly inhibited by osteoclasts, thus resulting in attenuated T-cell-mediated anti-myeloma immunity. Furthermore, the immunosuppressive effects of osteoclasts are alleviated, and IDO expression is decreased after CD38 monoclonal antibody is applied [23].
3. OVERCOMING STRUCTURAL COMPONENTS
The TME, apart from immunosuppressive cells, also includes structural components, including ECM and blood vessels. MM cells and cancer-associated fibroblasts synthesize and alter the ECM, while stimulating the growth of host blood vessels. In this way, the nature of TME is altered, which in turn promotes the growth and metastasis of MM.
3.1 ECM
As a major component of the MM TME, ECM is composed of different macromolecules, including proteoglycans, glycosaminoglycans, laminins, collagens, and their receptors in the TME [15]. It is essential for normal tissue homeostasis to maintain the integrity of the ECM network because the network provides structural support for the normal tissue and regulates cell morphology, function, and cell-matrix interactions [58]. MM cells modify the composition of the ECM via enzymatic and non-enzymatic processes, thereby promoting drug resistance, tumor cell survival, and proliferation. Eckhardt et al. [59] reported that transcription of ECM-related components is dysregulated during tumor progression through gene-expression profiling. In addition, TME and ECM are dynamic regulatory reservoirs for various effectors, including cytokines, growth factors, enzymes, and chemokines, which bind to glycosaminoglycans mainly via heparan sulfate side chains [58]. MM cells release ECM-bound effectors by secreting heparanase to cleaves heparan sulfate, which converts these effectors to bioactive mediators and regulates the composition and organization of the ECM [60]. In addition, MM cells interact directly with the ECM by binding syndecan-1 or very late antigen-4, thus leading to cell cycle dysregulation and activation of anti-apoptotic pathways within MM cells [61].
In addition to modification of the ECM, a permissive TME can form for MM cells. Of increasing interest is the formation of the pre-metastatic niche by MM cells by altering the ECM in the monoclonal gammopathy of undetermined significance (MGUS) stage [15]. Such an alteration arises from the deposition of ECM fragments, as well as new macromolecules by the enzymatic activity of malignant plasma cells [62]. Multiple ECM receptors, ECM proteins, and ECM-modulating enzymes, including laminin α4, integrin α5β5, and matrix metalloproteinase-2, have been reported to be upregulated in a step-by-step fashion from MGUS to MM by proteome profiling [63]. It is unclear, however, how the presence of the ECM affects the efficacy of CAR-T cell therapy. Peri-tumoral ECM collagen fibers have been reported to be a barrier that limit the access of T cells to the TME [64]. Therefore, infiltration of CAR-T cells into the TME can be promoted by matrix degradation, which in turn enhances the efficacy of CAR-T cell therapy.
3.2 Tumor vasculature
Under physiologic conditions, angiogenesis in the BM is regulated by numerous pro- and anti-angiogenic modulators, including cells, cytokines, and growth factors. The dynamic balance of angiogenesis is disrupted in the TME, and MM cells promote the growth of blood vessels by upregulated expression of pro-angiogenic modulators and downregulated expression of anti-angiogenic modulators, thereby leading to increased angiogenesis and tumor progression [65]. The level of pro-angiogenic factors or degree of angiogenesis in the MM TME is associated with the stage of disease, prognosis, and depth of response [66]. Furthermore, MM patients with active disease have a higher microvascular density compared to MGUS or inactive MM patients. A stepwise elevation in TME angiogenesis has been reported among patients with MGUS to smoldering MM, symptomatic MM, and relapsed MM [67]. Such elevated levels of angiogenesis in MM patients may be associated with the accumulation of pro-angiogenic cytokines, including VEGF, syndecan-1, and bFGF [67].
Aberrant angiogenesis is critical for the survival, proliferation, progression, and metastasis of tumor cells. Furthermore, aberrant angiogenesis provides a barrier that limits access and infiltration of CAR-T cells into the TME. To overcome the tumor vasculature, CAR-T cells can be designed to target pro-angiogenic modulators, including VEGF receptor (VEGFR)-1, VEGFR-2, prostate-specific membrane antigen (PSMA), or tumor endothelial marker 8 [40]. It has been shown that treatment with anti-VEGF antibodies results in increased recruitment of effector T cells and improved responses in metastatic renal cell carcinoma [68]. Corollary studies have demonstrated a reduced frequency of MDSCs in the TME after treatment with VEGFR-2-specific CAR-T cell therapy combined with IL-12, and showed promising efficacy in murine models with multiple vascularized tumors [69]. Additional studies are needed to target pro-angiogenic modulators in the TME and modify next-generation CAR-T cells to further enhance the efficacy of CAR-T cell therapies.
4. OVERCOMING HYPOXIA, NUTRIENT STARVATION, AND METABOLISM
Hypoxia and nutrient starvation are the main features of the MM TME. Hypoxia of the TME comes from the reduced oxygen concentration due to inadequate blood supply, increased oxygen consumption by MM cells, and hydrogen peroxide production [11, 15]. It has been shown that the oxygen concentration in the BM of MM patients or murine models is lower compared with healthy controls, which results from rapid cell division and aberrant angiogenesis [70]. Hypoxia can significantly increase the expression of CXCR4 in the TME, which ultimately promotes the homing and metastasis of circulating malignant plasma cells to other parts of the BM [71]. In addition, intra-tumoral hypoxia promotes epithelial-mesenchymal transition-like features of MM cells, and decreases E-cadherin expression and BM adhesion of MM cells, thereby enhancing tumor proliferation and migration [71]. To overcome the hypoxic TME, Juillerat et al. [72] designed a new type of hypoxia-induced factor (HIF) CAR, which fuses CD19-specific scFv and HIF1α sub-domains to γ and β chains of the IgE receptor. The function of such oxygen-sensitive CAR-T cells can be modulated by oxygen levels in the TME, paving the way for creating more “self-decision-making” CAR-T cells in the future.
Essential amino acids for T cell metabolism, such as such as tryptophan, are reduced in MM TME [73]. MM cells, MDSCs, and osteoblasts degrade tryptophan by overexpressing IDO, leading to suppressed proliferation of T cells. Furthermore, it has been shown that the expression of IDO in the TME also inhibits the immune effector activity of CAR-T cells, which results in failure of anti-CD19+ CAR-T cells to control the progression of CD19+ IDO-expressing tumors [74]. Therefore, the combination of inhibitors targeting IDO to modify the microenvironment while killing MM cells can be considered to maximize the efficacy of CAR-T cells.
The metabolism, differentiation, and function of T cells can also be modulated by metabolic stress from the TME. CAR-T cells with different co-stimulatory domains rely on different metabolic pathways. Choi et al. [75] showed that the 4-1BB co-stimulatory domain enhances the mitochondrial respiratory chain and catabolism in CAR-T cells. In contrast, the CD28 co-stimulatory domain favors the glycolysis pathway and lactate production [76].
5. CONCLUSION
Despite the significant efficacy achieved by CAR-T cell therapy, relapse following therapy has become one of the major problems for such a treatment paradigm. In addition to tumor progression and problems related to CAR-T cell products, TME is a determinant of the efficacy of CAR-T cell therapy in MM [77, 78]. In this review we summarized the main challenges for CAR-T cells in the TME, including immunosuppressive cells, structural components, hypoxia, nutrient starvation, and metabolism. We also proposed strategies to improve the efficacy of CAR-T cell therapy by modifying the TME.
CAR-T cell therapy, as ‘the living drug,’ is different from other immunotherapies because CAR-T cell therapy is more susceptible to the immunosuppressive TME that limits its efficacy. There is ample evidence that soluble factors, including IDO, IL-10, TGF-β [79], and immune checkpoints, such as PD-1 and CTLA-4 [80], cause T cell dysfunction. Furthermore, trafficking of CAR-T cells into the TME, aberrant tumor vasculature, and suppression of CAR-T cells due to immunosuppressive cells are some key challenges that the TME poses to CAR-T cells. In this review we summarized strategies for CAR-T cells to overcome the MM TME. These include depleting immunosuppressive cells, blocking immune checkpoint pathways, and modifying CARs to target the immunosuppressive cells and structural components in the TME. In addition, there are several other strategies under preclinical investigation, including alternative manufacturing processes [81], dual-target CAR-T cells, such as dual BCMA/CD19 CAR-T cells [82], and allogeneic CAR-T cells [83]. The ideal CAR-T cells for treatment in MM should meet the following characteristics: (1) target different MM cell sites to prevent antigen loss; (2) utilize the “self-decision-making” function to overcome the TME, while reducing toxicity and side effects; and (3) retain the naïve/memory phenotype to increase persistence. With the advances in gene editing technology and vector designs, better individualized and multi-functional CAR-T cells will be designed.