As mentioned above, the clinical efficacy of CAR-T cells observed in hematological malignancies is rarely found against solid tumors. Hematological malignancies are devoid of many of the physical immunosuppressive factors that hamper adoptively transferred cells from reaching solid tumors. Furthermore, target antigens that are present on hematological cancers are often homogenous and are expressed in most tumor cells. In contrast, target antigens on solid tumors are often heterogeneous both across tumors and between primary and metastatic sites. CAR-T cell therapy for solid tumors faces additional obstacles [
9,
10], starting from the administration route, wherein CAR-T cells injected in the peripheral blood must encounter the correct chemotactic signals to traffic to the tumor in sufficient numbers. The abnormal cancer vasculature impedes efficient passage (diapedesis) toward tumor tissues [
33,
34,
35]. Physical barriers by hard-to-penetrate fibrotic stroma prevent adequate CAR-T cell diffusion at a distance from the blood vessels. Finally, immunosuppressive factors include inhibitory checkpoint pathway signals and immunosuppressive cytokines [
36], TGF(transforming growth factor)-β among them [
37,
38,
39]. Products of an altered metabolism, including amino acids, lipids, and DNA base precursors [
40,
41,
42], and reactive oxygen species (ROS) [
43] can be found in the tumor environment [
12]. All these factors considerably affect the long-term persistence within the tumor in an active state.
3.1. CAR Target Choice for Anticancer Therapy
An appropriate choice of target antigens and the effective combination with immunostimulatory signaling have been shown to enhance CAR efficacy. CAR comprising the ICOS signaling domain liaises with the effective antitumor effect on gliomas that express the epidermal growth factor receptor variant III (EGFRvIII) [
44]. The preclinical evaluation of CAR-T cell therapy targeting the tumor antigen 5T4 in ovarian cancer led to successful outcomes [
45]. CAR targets included genetic products arising from gene mutations (
EGFRvIII) [
46], modified glycosylation patterns (MUC1) [
47], cancer-testis antigen-derived peptides (MAGE), and mesothelin-specific CAR-T cells [
48]. CAR was further generated that target overexpressed antigens in breast cancer, lung cancer, and pancreatic cancer, such as carcinoembryonic antigen GD2, prostate-specific membrane antigen, HER2/ERBB2, MUC16 [
49] or tumor stroma (fibroblast activation protein or vascular endothelial growth factor receptor, VEGFR) [
50]. Additional novel targets include the type 1 insulin-like growth factor receptor (IGF1R) and receptor tyrosine kinase-like orphan receptor 1 (ROR1) for sarcoma as well as the L1-cell adhesion molecule (L1-CAM) for ovarian cancer [
51].
These combined efforts have led to a burgeoning of clinical trials in the field, the majority remaining focused, though, on leukemias and lymphomas [
12]. FDA-approved CAR-T cell therapies now include CTL019 (Kymriah) [
52], KTE-C19 (Yescarta) [
53], JCAR017 (Breyanzi) [
54], KTE-X19 (Tecartus) [
55], and bb2121 (Abecma) [
56].
3.3. CAR-T Treatment Toxicity
The anticancer activity of CAR-T can associate with life-threatening toxicity due to the increased secretion of pro-inflammatory cytokines (cytokine release syndrome, CRS) and the immune effector cell-associated neurotoxicity syndrome (ICANS) as mediated by the simultaneous activation of large numbers of T cells [
8,
58]. This frequently leads to multiorgan dysfunction, pulmonary failure, and death [
59]. Consequent therapeutic intervention requires the intense management of treated patients [
60,
61].
Cytokine release is usually greater with CAR containing CD28 versus 4-1BB co-stimulatory domains. On the other hand, constructs with either domain confer similar anticancer activity in mouse models. T cell products expressing CAR with either CD28 or 4-1BB co-stimulatory domains have been highly efficacious in patients with relapsed hematological malignancies, and anti-CD19 CAR showed similar activity regardless of the source of the co-stimulatory domain. In large-cohort clinical trials, the rates of neurological toxicities have been higher with CD28–co-stimulated CAR, although this finding is probably the result of multiple converging factors rather than due to CD28 signaling alone [
62]. One of these factors was the increased circulating IL-17 levels at baseline in patients with locoregional metastatic melanoma. In ICB approaches, increased IL-6 levels in patients with metastatic melanoma treated with ipilimumab (anti-CTLA-4 antibody) were shown to be associated with more intense adverse events. More global cytokine dysregulation as assessed by measuring the circulating levels of several cytokines at baseline or early on treatment has been shown to be predictive of adverse events in patients treated with anti-PD-1 therapies alone or in combination with anti-CTLA-4 therapies [
21].
Intense cytokine release is associated with T-cell activation upon engagement with target cells, which leads to higher levels of circulating cytokines, including IL-6 and interferon γ. Consistently, elevated levels of serum cytokines are less common in patients who do not have a clinical response after CAR-T cell therapy [
63].
NK cells produce several cytokines, including tumor necrosis factor α, interferon γ, and IL-10 [
64]. NK cells produce lower levels of IL-6 than T cells [
63], thus potentially reducing systemic cytokine-storm toxicity. Of note, the CAR-T cell-induced cytokine release syndrome can be mediated by macrophages and is relieved by IL-1 blockade [
65].
3.4. CAR-T Cell Exhaustion
A major concern of CAR-T therapy is that CAR-T cells may become exhausted or dysfunctional [
66,
67], which is a phenomenon whereby the T cells become unresponsive due to overstimulation [
68].
CAR-T cell exhaustion is considered the outcome of the chronic tumor stimulation imposed on T cells, which leads to disruption of their function [
67]. Proof of (re-)activation of TIL upon blockade of the PD-1/PD-L1 (programmed death-ligand 1) or other checkpoint axes [
69] has strongly supported this model. Correspondingly, other therapeutic measures, including adoptive T-cell therapy, epigenetic reprogramming, antibodies targeting T-cell co-stimulatory molecules, metabolic reprogramming, and conventional cancer therapies, such as chemotherapy, radiotherapy, and targeted therapy, have been implemented to enhance antitumor immunity, overcome resistance, and increase therapeutic efficacy [
22].
Cell exhaustion may differentially affect T cell subpopulations, suggesting a relationship with the selection procedures utilized to manufacture CAR-T cells. Optimal cell subpopulations for adoptive cell transfer were suggested to be those that retain their memory/naïve capacities [
70] to permit a greater boost in proliferation and function in vivo. Wnt signaling was shown to promote the generation of CD44(low)CD62L(high)Sca-1(high)CD122(high)Bcl-2(high) self-renewing multipotent CD8(+) memory stem cells with proliferative and antitumor capacities exceeding those of central and effector memory T cell subsets [
70].
Epigenetic profiles regulate the gene expression of key transcription factors over immune cell differentiation and proliferation pathways. Through a screening of chemical probes with defined epigenetic targets, JQ1, an inhibitor of bromodomain and extra-terminal motif (BET) proteins, was found to maintain CD8+ T cells with functional properties of stem cell–like and central memory T cells [
71]. Adoptively transferred in vitro-JQ1-treated CAR-T cells showed higher proliferation, persistence, and increased cytokine secretion than non-treated CAR-T cells in murine models [
71].
The altered differentiation of CAR-T cells can also accompany T cell exhaustion [
72]. Utilizing mesothelin-redirected CAR-T cells in pancreatic cancer, CAR dysregulation was found to be associated with a CD8+ T-to-NK-like T cell transition, as driven by SOX4 (SRY-Box Transcription Factor 4) and ID3. The downmodulation of ID3 and SOX4 expression was indicated to improve the efficacy of CAR-T cells in solid tumors by preventing or delaying T cell exhaustion [
72]. These findings further reveal CAR-T cells’ plasticity as a main actor of immune response dynamics.
3.5. Counteracting Immunosuppression against CAR-T Cells
A major hurdle to be overcome by tumor-infiltrating CAR-T cells is immunosuppression. T cell exhaustion itself was shown to be an outcome of cancer-associated immunosuppression. Correspondingly, resistance to exhaustion was shown to be linked to interaction with other cancer-infiltrating cell types [
12].
Cancers contain a broad ensemble of genetically normal cells within an extracellular matrix (ECM), which was collectively termed the tumor microenvironment (TME), that substantially diverges from normal stroma. This has fostered the concept that tumors grow as integrated tissues or organs, that combine diverse components, such as vasculature, nerves, an immune environment, and connective tissue.
TGF-β is a key driver of immunosuppression [
37,
38,
39]. Prostate cancer, in particular, secretes TGF-β as a means to inhibit immunity while allowing for cancer progression. Blocking TGF-β signaling augments T cell ability to infiltrate, proliferate, and mediate antitumor responses in prostate cancer models. The potency of PSMA(prostate-specific membrane antigen)-targeted CAR-T cells was correspondingly enhanced utilizing dominant-negative TGF-βRII (dnTGF-
βRII) expression in CAR-T cells [
73]. This led to an increased proliferation of CAR-T cells, enhanced cytokine secretion, resistance to exhaustion, long-term in vivo persistence, and eradication of human prostate cancer in mouse models. A phase I clinical trial is being conducted to assess these CAR-T cells in relapsed and refractory metastatic prostate cancer patients. This is a non-randomized, sequential assignment, open label trial; 18 participants have been enrolled; completion of the study is expected in 2022 (
ClinicalTrials.gov: NCT03089203) [
73]. Knocking out the endogenous TGF-β receptor II (TGFBR2) in CAR-T cells was achieved using CRISPR/Cas9. This reduced Treg conversion prevented CAR-T cell exhaustion and achieved higher tumor eradication rates both for xenografts and for PDX (patient derived xenograft), with higher proportion of memory and effector memory CAR-T cell subsets [
74]. Additional approaches were suggested to be effective, e.g., the expression of chimeric TGFBR2 and TGFBR1 where the TGF-β-binding domain is fused to the transmembrane and intracellular signaling domains of IL-12 receptor (CTBR). CAR-T/CTBR cells secreted significantly greater amounts of IFNγ than control T cells following activation in the presence of TGF-β. In the absence of IL-2, antigen-driven expansion was severely limited by exposure to TGF-β, and CAR-T cells progressively lost cytotoxic activity. Although T cells overexpressing the dominant negative TGF-β receptor failed to expand and clear tumor cells in the presence of TGF-β, CTBR expressing CAR-T cells maintained their ability to expand and kill tumor targets in the presence of TGF-β [
75].
Metabolic conditions can negatively impact on CAR-T cell function: among them, an increase in the acidity of the TME because of increased glycolysis by cancer cells. This ‘Warburg effect’ [
76,
77] stems from a preferential utilization of glucose via glycolysis rather than via oxidative phosphorylation. Cancer-associated fibroblasts (CAF) contribute to increased intratumor glycolysis and impact on breast cancer growth [
78]. High glycolysis leads also to an increase in oxidative stress and in ROS production. The secretion of high levels of ROS by MDSC contributes additional immunosuppressive capacity. Immunosuppression by oxidative stress impairs CAR-T cells proliferation and cytotoxicity [
12]. This led to engineering CAR-T cells to secrete catalase (CAT), an antioxidant enzyme, into the TME. CAR-CAT-T cells were shown to regain their antitumor functions [
43]. Local catalase secretion provided a bystander effect and restored cytotoxic function to NK cells.
One of the approaches used to fight immunosuppression has been the generation of CAR-T cells expressing cell-surface dominant-negative receptors (DNR) to override the inactivating signals present in the TME. DNR can be generated with a functional extra-cellular domain and a mutation in the intracellular region to abolish downstream signal transduction. DNR can effectively compete with their endogenous counterparts. The use of DNR for immunosuppressive factors such as TGF-β has endowed transduced EBV cells with resistance to immunosuppression [
79]. A DNR for PD-1 on CAR-T cells rescued the effect of checkpoint blockade and restored effector functions. PD-1/PD-L1 blockade is normally achieved through systemic antibody delivery, which can result in autoimmune reactions. PD-1 ‘insensitive’ DNR T cells do not require systemic ICB and may prevent this major side effect. Switch receptors offer yet another approach to circumvent immunosuppression. These CAR contain the extracellular portion of an antibody specific for an immunosuppressive molecule, such as PD-1 or CTLA-4, which is fused to an intracellular activating signaling molecule, such as CD28. The infiltration and antitumor efficacy of PD-1-CD28 switch-CAR-T cells were enhanced versus parental CAR-T cells [
80]. Switch-CAR-T cells showed a reduction in other checkpoint inhibitors, e.g., LAG3, TIM-3, and CEACAM1 (carcinoembryonic antigen-related cell adhesion molecule 1), and increased IL-2 signaling, suggesting an induction of recovery from cell exhaustion.
The reduction of inhibitory signaling pathways in T cells has shown promise in T-cell reactivation. The inhibition of Protein Kinase A with Ezrin using a ‘regulatory subunit 1 anchoring disruptor’ (RIAD-CAR) resulted in an upregulation of CXCR3 and CD49D integrin (VLA-4), which resulted in enhanced RIAD-CAR-T cells trafficking to tumors and better migration to CXCL10 in vitro [
81]. RIAD-CAR cells expressed higher levels of both IFN
γ and cytotoxicity and were more resistant to immunosuppression in TME.
IL-8 release within tumors was utilized to enhance intratumoral T-cell trafficking. Modified CAR inducing the expression of IL-8 receptors, CXCR1 or CXCR2, showed enhanced migration and persistence of T cells in the tumor. This induced complete tumor regression and long-lasting immunologic memory in preclinical models of glioblastoma, ovarian, and pancreatic cancer [
11].
TGF-β-resistant EGFRvIII CAR-T cells were shown to possess higher antitumor efficacy in murine glioma models [
82], and prolonged survival was observed following EGFRvIII CAR-T cell treatment in advanced glioblastoma [
83].