712
views
0
recommends
+1 Recommend
1 collections
    1
    shares

      Interested in becoming a HOD published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      See further information on submitting a paper at https://hod-journal.org/submit-a-paper/

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      T-cell engineering strategies for tumors with low antigen density, and T-cell survival in the immunosuppressive tumor microenvironment of relapsed/refractory diffuse large B-cell lymphoma

      Published
      review-article
      Bookmark

            Abstract

            Refractory and/or relapsed (r/r) diffuse large B-cell lymphomas after treatment with two lines of systemic chemoimmunotherapy exhibit diversity in genetics, tissue biology, and pathology, as well as poor prognosis. Patient TCRαβ cells engineered with a CD19-specific chimeric antigen receptor (CAR) have shown promising clinical outcomes in r/r diffuse large B-cell lymphoma. The ZUMA-1 study, the JULIET study, and the TRANSCEND NHL 001 study of three prototype 19CAR-T cells have indicated an overall response rate of 52–82%, a complete response rate of 40–58%, and a 12-month progression-free survival of 33.2%–46.6%, with clinically manageable treatment related toxicity. At the 5-year follow-up, relapse was observed in approximately 57% of patients within 1 year. Understanding of the risk factors for non-response remains insufficient. In addition to intrinsic tumor resistance, such as aberrant apoptotic signaling, downregulation or loss of tumor-associated antigens (TAA), an immunosuppressive tumor microenvironment, and CAR-T cell exhaustion in vivo have been suggested to be important risk factors. Mechanisms underlying 19CAR-T cell exhaustion under chronic TAA exposure, and limited 19CAR-T cell trafficking and infiltration into the tumor mass have been reported. Moreover, tumor escape in the presence of low TAA density remains a challenge in 1928ζ CAR-T cell treatment. In this review, we provide an overview of modified modular CAR elements and their synergistic effects in controlling T-cell function. We then briefly discuss novel strategies against tumors with low TAA density, such as bispecific tandem or loop CAR recognition domains, the development of human leukocyte antigen-independent synthetic TCRαβ double-chain receptors integrated into the constant region of the TCRα chain, and armored CAR-T cells targeting the tumor microenvironment.

            Main article text

            1. INTRODUCTION

            Lymphoid neoplasms are broadly divided into B-cell lymphoid, T-cell, NK-cell, and dendritic cell neoplasms, and Hodgkin lymphomas (HL) [13]. In contrast to the cure rate of ≥ 85% in patients (pts) with HL [46], the morbidity and mortality of adult non-Hodgkin lymphoma (NHL), including 45.8% of diffuse large B cell lymphoma (DLBCL), ranked 10th death in China before 2016 [7, 8]. Among more than 18 different DLBCL subtypes, worse outcomes were reported with co-occurrence of MYD88 L265P and CD79B mutation in activated B-cell-like (ABC)-DLBCL (MCD subtype), and ABC-DLBCL with gain-of-function of NOTCH1 mutation (N1 subtype). MCD and N1 subtypes had 26% and 36% 5-year overall survival (OS) after rituximab-based chemoimmunotherapy, respectively [9]. In addition, high-grade-B cell lymphomas with c-Myc/8q24 and BCL2/18q21 translocation (double hit) or with BCL6/3q27 translocation (triple hit) are highly resistant to chemotherapy [10]. Before the advent of autologous CD19 chimeric antigen receptor (CAR)-T cell (19CAR-T cell) therapy, the 2-year OS of patients with chemotherapy-resistant DLBCL was 20%, and the median OS was 6.2 months, according to the SCHOLAR-1 study [11]. Axicabtagene ciloleucel (axi-cel), prototypic 1928ζ CAR-T cells, have been reported to result in a 54% 2-year OS in patients with r/r DLBCL after two or more lines of treatment in the ZUMA-1 study [12, 13]. In addition, the introduction of axi-cel into second-line therapy in patients with r/r DLBCL or infusion of axi-cel as part of first line therapy in patients with double- or triple-hit HGBCL significantly increased event-free-survival (EFS), and overall response rate (ORR)/complete response (CR) compared with standard therapy [14, 15].

            Despite high treatment effectiveness, grade ≥3 cytokine release syndrome (CRS) and immune cell associated neurotoxicity (ICAN) from axi-cel occur in 13% and 28% of treated patients with r/r DLBCL, respectively, after two or more lines of treatment [1618]. Notably, CRS increases rapidly within 5–10 days after transfusion of axi-cel [17]. The clinical and biological factors of individual patients have been associated with the risk of CRS and ICAN after 19CAR-T cell therapies. These factors include individual inflammatory status, intestinal microbiota, tumor burden, genomic instability of r/r DLBCL, lymphocyte depletion protocols, intrinsic quality characteristics of 19CAR-T cells, pyroptosis of B-cell cancer, endothelial or macrophage activation, and IL-6 level [1923]. During CRS progression, local (L)-CRS is observed in patients with a high tumor burden, including local activation and expansion of 19CAR-T cells, activation of myeloid cells [24], and secretion of tumor necrosis factor (TNF)-α, interferon γ, and IL-1ß. L-CRS is a precursor of systemic (S)-CRS [25]. S-CRS is ameliorated by synergistic blockade of TNF-α, IL-1ß, and inhibition of focal adhesion kinase (FAK) [26]. In addition, tocilizumab, corticosteroids, and ruxolitinib are frequently used to control S-CRS [2729]. In addition, metoprolol, a cardioselective ß1-adrenergic receptor blocker, has recently been reported to improve CRS and serum levels of IL-6 [30].

            Approximately 57% of patients with r/r DLBCL experience disease progression or relapse after autologous 19CAR-T cell therapy [31, 32]. For example, early disease progression within 30 days or after 30 days has been reported to have a median OS of 3.75 months and 9.28 months, respectively [33]. Tumor intrinsic factors, such as two or more extra nodal sites, a high total metabolic volume, elevated serum C-reactive protein (CRP) and lactate dehydrogenase, the presence of apolipoprotein B mRNA editing catalytic polypeptide 3 (APOBEC), cytidine deaminase mutagenesis-associated signatures 2 and 13, the presence of 3p21.31, a chromosomal deletion containing the tumor suppressor RHOA, aberrant apoptotic signaling, and oxidative DNA damage are closely associated with early disease progression after 19CAR-T cell treatment [34, 35]. In addition, CD19 loss and/or decrease in B-cell cancer is associated with resistance 19CAR-T cell therapy [36]. The mechanisms of CD19 loss in B-cell malignancies have been extensively studied [36]. These mechanisms include pre-existing CD19-negative clones [37], acquired genetic point mutation of CD19 exon 3 from p.163 (R to L) or p.174 (L to V) [38], loss of serine/arginine-rich splicing factor 3 (SRSF3) [39], hyper-glycosylation of CD19 [40], myeloid lineage switch due to dysregulation of PAX5 [41], Ikaros DNA binding protein [42], and the EBF1 zinc finger containing transcription factor [43]. Moreover, the presence of fewer than 3000 membrane bound CD19 molecules per r/r DLBCL results in failure to activate 1928ζ CAR-T cells. This aspect is reflected by decreased phosphorylation of extracellular signal-regulated kinase (pERK) in 19CAR-T cells [44, 45]. Furthermore, 19CAR-T cell exhaustion under an immunosuppressive tumor microenvironment (TME) leads to disease progression or relapse. Chronic TAA exposure in the TME results in constitutive activation of 19CAR-T cells, differentiation, epigenetic alteration, and upregulation of immune checkpoint molecules [4648]. Recently, numerous novel strategies have been developed to modify T cells to overcome TME and tumor heterogeneity TAA. This literature review provides an overview of modular models of prototypical CAR constructs, and their synergistic effects in controlling T-cell function. Subsequently, novel CAR modifications and T-cell engineering strategies are summarized. Notably, bispecific tandem or loop CAR-T cells significantly enhance therapy-associated safety and high efficacy in patients with r/r DLBCL. In addition, CAR integrating at TCRα chain constant region by using clustered regularly interspaced short palindromic repeats (CRISPR) derived single guide RNA and Cas9 and recombinant adeno associated viral vectors (rAAV) enhances tumor rejection, and delays CAR-T cells exhaustion in vivo. Furthermore, human leukocyte antigen independent synthetic TCR and antigen receptors are able to respond to low density tumor antigens with little tonic signaling. Armored CAR-T cells facilitate CAR-T cell migration and infiltration, and survive in the TME of r/r DLBCL.

            2. CAR ELEMENTS AND THEIR SYNERGISTIC EFFECTS IN CONTROLLING CAR-T CELL FUNCTION

            2.1 Affinity and avidity of CAR

            The B cell response to antigens depends on the antigen/BCR affinity threshold [49]. Activation of TAA specific CAR-T cells depends on the affinity of CAR, surface CAR density, and TAA expression per cancer cell [5052]. Typical second-generation CAR constructs have had great success in treating hematologic cancers [5355]. Second-generation CARs consist of five modular domains: the extracellular TAA binding domain, ectodomain of the hinge region, transmembrane domain, co-stimulatory domain, and three repetitive immune tyrosine activation motifs (ITAM) of the CD3ζ chain. The ectodomain of the antigen binding domain, together with the hinge region, facilitates TAA recognition. The second-generation CAR construct was developed from the first-generation antibody-based CAR construct, designed in 1989. The single chain variable fragment (scFv) of mouse monoclonal antibodies (mAb) against TAA is commonly used as a TAA binding domain. The development of camel heavy-chain antibody, human Fab antibodies, and natural immune receptors, such as NKG2D, has expanded the number of disease targets [56]. At the molecular level, scFv consists of a variable heavy chain (VH) and a variable light chain (VL). Each VH and VL has four framework regions (FR1–4) and three complementarity determining regions (CDR1–3). Specifically, CDR1–3 of CAR-scFv (FMC63 mAb, IgG2a) bind membrane-bound epitopes adjacent to exons 3 and 4 of CD19 [28]. FR1–4 of CAR-scFv facilitate binding of CDR1–3 to CD19. Studies have shown that FR1–4 of scFv of FMC63 mAb are much more stable than FR1–4 of scFv of GD2 specific CAR-T cells and CD22 specific BBζ CAR-T cells. Therefore, 19BBζ CAR-T cells have little antigen-independent scFv-clustering that triggers tonic signaling. In general, CAR-T cell tonic signaling results in TAA independent constitutive CD3ζ activation, terminal differentiation, and exhaustion of CAR-T cells [57, 58]. However, tonic signaling delivers activation signals to CD22BBζ CAR-T cells (m971 mAb) and overcomes insufficient reactivity against low-density CD22 TAA in r/r B-ALL. For CD22BBζ CAR-T cells, short scFv linkers (5 amino acids) have a much more autonomous tonic signaling effect through antigen-independent multimerization of scFv than the long linker sequence of 20 amino acids. The affinity of CD22BBζ CAR-scFv with a short-linker sequence (6.10 nM) is not significantly different from that of CD22BBζ CAR-scFv with a long-linker length (1.70 nM) [59, 60]. The affinity of scFv can be measured with the equilibrium dissociation constant (KD) toward monovalent TAA in solution. The KD value and affinity of CAR-scFv of mAb are inversely related. The affinity of CAR-scFv is often considered an important parameter for evaluating the specificity and sensitivity of TAA. The affinity of scFv of mouse mAbs ranges from 1 micromolar (μM) to 1 nanomolar (nM). To date, no guidelines are available for developing optimal CAR affinity for a given antigen. However, relevant retrospective studies evaluating clinical and preclinical data on the affinity of CAR for solid cancer cells have shown that an intermediate affinity (KD of ~20–100 nM) of CAR-scFv has better clinical efficiency than a higher affinity [50, 61]. However, this finding does not apply to the Food and Drug Administration (FDA)-approved 19CAR binding domain. The scFv of FMC63 mAb has been used in three prototypic 19CAR-T cells and has high affinity (KD of 0.33 nM) [57, 58]. A novel CD19 CAR-scFv of CAT mAb has 42-fold lower affinity (KD of 14 nM) than that of CAR-scFv of FMC63 mAb. Although both share a similar binding epitope, the relatively lower affinity of CAR-scFv of CAT mAb results in lower activation-induced cell death (AICD), CD19 antigen trogocytosis, or antigen reduction after CD19 binding. Moreover, persistence of the low-affinity of 19CAR-scFv of CAT mAb were superior compared to high affinity 19CAR-T cells in pediatric patients with r/r B-ALL [62, 63]. The low-affinity of CAR-scFv of CAT mAb decreases severe off-target toxicity [64]. In contrast to monovalent affinity, polyvalent interaction between multiple receptors, particularly dimerized CAR and ligands, increases the overall binding strength (functional avidity) and thus antigen sensitivity. For example, CAR dimerization by two cysteine residues of the proximal region of the CD8α transmembrane domain (TMD) increases the functional avidity of CAR-T cells. Moreover, the CD8α TMD of CAR-T cells stabilizes the surface expression of CAR in T cells [65, 66]. The extracellular location of the TAA, and its accessibility for binding CAR-T cells is determined by the hinge region of the CAR ectodomain. Post-translational modification by N-linked glycosylation of the CD28 hinge domain facilitates CAR surface expression [67]. Moreover, CAR using the CD28 hinge forms a homodimer through its cysteine residue. Furthermore, CD28 TMD of CAR facilitates the formation of a heterodimer with the endogenous CD28 molecule of T-cells through the conserved YxxxxT motif [68]. Axicabtagene ciloleucel contains a CD28 hinge, CD28 TMD, and CD28 co-stimulatory domain, and thus has higher TAA sensitivity than FDA-approved lisocabtagene maraleucel (Liso-cel) and tisagenlecleucel (Tis-cel) ( Figure 1 ) [52, 6971]. The immunoglobulin (Ig) like domain of the CD8α hinge and TMD (~5 nm) of Tis-cel; Ig like domain of CD28 hinge and TMD (~6 nm) of axicabtagene ciloleucel (axi-cel); and mutant IgG4-based long hinge and CD8α TMD (~7 nm) of Liso-cel show a non-classical immune synapse structure after ligand binding [12, 52, 68, 71, 72]. The spatial boundary of the immune synapse is approximately 15 nm. According to the segregation model of T cell activation, use of a proper hinge size (<15 nm) of CAR-T cells efficiently excludes the negative regulator of phosphatase CD45 from the non-classical immune synapse before antigen-dependent phosphorylation signals are transmitted by kinases [73, 74]. Although the CD8α hinge resists hinge proteolysis, modification of the hinge length (39 amino acids) decreases the treatment-associated adverse effects of Tis-cel with 19BBζ (71) CAR-T cells. Notably, the long CD8α hinge length of 55 aa in 19BBζ (86) CAR-T cells is associated with a diminished incidence of severe CRS and ICAN in patients with r/r B-NHL, but promising clinical efficacy [69, 75].

            Figure 1 |

            Functional representation of three prototypical FDA-approved CD19-specific CAR elements, and their delivery viral vectors, promoters, and cellular starting material for autologous CAR-T cell production.

            2.2 Costimulatory domains for CAR

            Incorporation of the costimulatory domain 4-1BB (BB) results in more than 1000-fold in vivo expansion and 10-year long-term persistence of CD4+19BBζ CAR-T cells in patients with chronic lymphocytic leukemia (CLL) [76, 77]. In contrast, first-generation 19ζ CAR-T cells lacking the co-stimulatory domain have an in vivo persistence of 7 days in patients with LBCL, and human anti-mouse CAR-scFv has been reported; this treatment does not produce potent anti-tumor effects [7881]. After activation, the cytoplasmic domain of 4-1BB constitutively potentiates non-canonical NF-κB signaling, thereby downregulating Bim apoptotic proteins, up-regulating anti-apoptotic B cell lymphoma-extra-large (Bcl-xL), and activating ERK of 19BBζ CAR-T cells [82]. Moreover, uptake of 4-1BB in CAR promotes mitochondrial biogenesis, mitochondrial fatty acid oxidation, and metabolic reprogramming of central memory CAR-T cells. Faster in vivo expansion of CD8+19CAR-Tcm cells than CD4+19CAR-Tcm cells has been observed [83, 84]. In addition, antigen independent CAR-scFv self-aggregation and tonic 4-1BBCD3ζ signaling continually activate TRAF2-NF-κB signaling and Fas dependent cell death [85]. Toxicity to T cells can be mitigated by attenuating CAR expression through the self-inactivating non-long terminal repeat (LTR) promoter of a lentiviral vector [86]. Moreover, 4-1BB co-stimulation ameliorates the exhaustion of 19BBζ CAR-T cells compared with 1928ζ CAR-T cells [58]. Furthermore, 19BBζ CAR-T cells ameliorate exhaustion by recruiting THEMIS-SHP1 phosphatase, which decreases the phosphorylation of CAR-CD3ζ [87]. Moreover, 19BBζ CAR-T cells have slower tumor rejection kinetics than 1928ζ CAR-T cells after initial antigen binding [88]. Intracellular signaling strength balances the effector function and memory development of CAR-T cells. Incorporation of the CD28 tyrosine residue YMNM-PYAP” proximal to, and CD3ζ distal to, the cell membrane facilitates binding of the p85 subunit of phosphatidylinositol 3-kinase (PI3K) and IL-2 secretion [89]. Activation of the PI3K-Akt-mTOR-PKC-theta-NF-κB pathway regulates glucose metabolism and T cell differentiation. In addition, adaptor proteins such as IL-2-inducible T-cell tyrosine kinase (Itk), filamin A, Lck, and Grb2/Vav interact with the distal carboxy-terminal proline-rich PYAP motif of the co-stimulatory domain of CD28. The involvement of the Slp-76/linker for activated T cell (LAT) proteins is less than that in native TCR signaling [90, 91]. Several downstream effectors lead to transcriptional activation of the canonical activated protein 1 (AP1) c-Jun, and Ca2+ flux-mediated calcineurin-nuclear factor of activated T cells (NFAT) pathway [92]. Thus, 1928ζ CAR-T cells with effector memory (EM) exhibit greater amounts of phosphorylated proteins and cytotoxic molecules, and faster in vivo tumor rejection kinetics than 19BBζ CAR-T cells during antigen recognition [93]. Percentage of EM phenotype in the final products of 1928ζ CAR-T cells are demonstrated to be one of key factors that positively affect complete clinical response in r/r DLBCL patients [94]. Phospho-proteomic analysis of activated 1928ζ CAR-T cells revealed a large abundant intracellular phosphorylation signals, which resulted in activation-induced cell death [87, 95, 96], because the in vivo proliferation kinetics and long-term persistence of CAR-T cells are closely correlated with the duration of clinical remission [97]. Genetic methods have been developed to balance the effector or memory function of 1928ζ CAR-T cells. For example, engineering of 1928ζITAM CAR-T cells to reserve an ITAM of CD3ζ proximal to the T cell membrane prolongs the in vivo persistence of 1928ζITAM CAR-T cells. In contrast, the intensity of intracellular signaling and antigen sensitivity is significantly attenuated [98]. In addition, substitution of CD3ζ in 1928ζ CAR-T cells with a CD3ɛ ITAM and CD3ɛ signal sequence decreases signal intensity and cytokine production through recruitment of the inhibitory Csk kinase [99]. Furthermore, genetic mutation of asparagine to phenylalanine in YMNM in CD28 decreases terminal differentiation of EM cells and exhaustion of 28ζ CAR-T cells [100].

            Third-generation CAR-T cells have been developed by selecting and positioning one or more co-stimulatory domains from members of the CD28/B7 IgG superfamily, and/or members of the tumor necrosis factor receptor type II (TNFR II) family, such as OX40, CD27, and CD40 [101]. Recently, an alternative type III transmembrane protein, B cell-activating factor receptor (BAFF-R), has been identified as a new potent costimulatory domain of CAR-T cells [102]. Direct fusion of CD28 and the 4-1BB costimulatory domain of 1928BBζ CAR-T cells has more favorable in vivo persistence in patients with minimal r/r NHL after autologous HSCT than 1928ζ CAR-T cells [103, 104]. Similarly, direct fusion of inducible T-cell co-stimulator (ICOS) proximally, and 4-1BB distally, from the cell membrane prolongs the survival and in vivo persistence of 19ICOSBBζ CAR-T cells [105]. In contrast to the direct infusion of two co-stimulatory domains, use of parallel CAR constructs for co-expression of membrane proximally positioned CD28 and 4-1BB results in functional persistence and enhanced anti-tumor rejection [106]. Moreover, parallel 1928ζ CAR constructs co-expression of membrane proximal 4-1BB ligand (L) not only support 1928ζ-41BBL persistence but also activate interferon regulatory factor 7 dependent type I interferon production. The release of IFNß further activates innate and adaptive anti-tumor immunity [107]. In another well studied case, parallel 20BBζ CAR incorporation of full-length antigen independent OX40 into T cells recruits TNR associated factors 2 and 5, thereby activating the NF-κB pathway, the anti-apoptotic gene BCL-2, and the PI3K-AKT pathway. The combination of 20BBζ CAR signaling and antigen independent full-length OX40 costimulatory signaling reduce 20BBζ-OX40 CAR-T cells apoptosis, exhaustion, and improved in vivo persistence, anti-lymphoma efficacy compared with 20BBζ CAR-T cells [108].

            A novel multiple chain chimeric immunoreceptor construct composed of ITAM with activating protein of 12 Da (Dap12) covalently binding the transmembrane domain of killer immunoglobulin-like receptor (KIR)S2, and KIRS2 conjugated to an antigen-specific scFv have been investigated. Notably, the antigen-specific KIRS2/Dap12 has stable surface expression and potent anti-tumor activity. After ligation of the antigen, KIRS2/Dap12 binds Syk and Zap70 kinase [109]. Optimization of CD19-scFv-KIRS2/Dap12-4-1BB engineered T cells by adding the co-stimulatory domain 4-1BB has been found to result in 100% CR and optimal IL-2 secretion, faster proliferation and persistence, and potent anti-tumor activity, as compared with non-co-stimulated CD19-scFv-KIRS2/Dap12. In addition, no ICANs and CRS have been detected in four adult patients with r/r B-ALL [110].

            2.3 CAR delivery with viral vectors

            To date, most autologous CAR-T cells and universal allogeneic CAR-T cells are produced ex vivo. The human immunodeficiency-derived third-generation self-inactivation lentiviral vector (SIN) with the U3 region of the 3´ long terminal repeat (LTR) removed minimizes the risk of replication-competent recombination. SIN LTR lentiviral vector constructs containing a central polypurine Tract (cPPT) sequence increase the transduction rate [111]. However, integration of CAR by lentiviral vectors at high multiplicity of infection results in multiple copies of CAR per T-cell genome and genotoxicity of the viral vectors. The FDA recommends that the viral vector copy number per CAR-T cell be fewer than five copies [112]. However, maintaining constitutive CAR transcription and protein expression with fewer than five copies per CAR-T cell is challenging. The selection of robust internal enhancer and promoter sequences can overcome epigenetic silencing, in addition to the introduction of a Woodchuck hepatitis virus post-transcriptional regulatory element in the 3´ untranslated region of the lentiviral vector of SIN LTR, the incorporation of a chromatin domain insulator flanking the properly sized transcription unit, and the addition of scaffold/matrix attachment regions [113116]. The murine stem cell virus-based γ-retroviral vector is used to make axi-cel, and the murine stem cell virus LTR controls the transcription of axi-cel. Intron-containing promoters such as Elongation Factor 1α are used to drive transcription of CAR in lentiviral vectors. Despite the efficiency of viral vector delivery [117], the immunological barrier of the cytosolic DNA sensor to the viral vector sequence, the epitope encoded by the viral vector, the immune response to the transgene, and the transcriptional repression of the viral vector remain challenges [118121]. In addition, the distribution of the lentiviral vector (LV) integration sites influences clinical treatment outcome by modulating in vivo proliferation of 19CAR-T cells in patients with chronic lymphocytic leukemia (CLL). Specifically, integration into the host T-cell methylcytosine dioxygenase TET2 allele together with a hypomorphic mutation in the second TET2 allele resulted in TET2 gene mutation. A single clonal proliferation of TET2-disrupted 19CAR-T cells with a less differentiated central memory phenotype induced CLL remission [119, 122, 123].

            2.4 Precision genome engineering of CAR

            Site-specific integration of CAR into the host genome has demonstrated clinical safety and efficacy. This method relies on the introduction of a site-specific break in the genome and the presence of homologous DNA sequences at double-stand breaks to perform high-fidelity homologous recombinational repair in host cells. In mammalian cells S or G2 phase, site-specific DNA double-stand breaks (DSB) could be generated by nucleofection of clustered regularly interspaced short palindromic repeats (CRISPR) associated system (Cas)9 and specific CRISPR Cas9 single guide (sg) RNAs efficiently and relatively precise. CRISPR Cas9/sgRNA is derived from the type II CRISPR/Cas system of microbial nuclease system. Moreover, rational design of the length and sequences of homologous arm of donor template DNA is key to HRR-mediated genome editing. Methods of delivering homologous donor template DNA include chemical transfection, pseudovirus particles from biological methods, and physical methods. Ex vivo electroporation, recombinant adeno associated virus (rAAV) donor vectors, non-integrating lentiviral vectors are efficient in transferring transgene incorporating donor template DNA [124, 125]. Another non-viral transgene approach for site-specific insertion of transgenes is the fusion of a highly soluble Sleeping Beauty (SB) 100X transposase with a catalytically dead Cas9 (dCas9). The choice of integration sites of SB 100X-dCas9 depends on the design of the gene-specific sgRNA. SB 100X transposon gene insertion sites are located approximately 300 base pairs downstream of sgRNA targets [126, 127]. Specifically, non-viral PD-1 locus integrated CD19-4-1BB-CD3ζ CAR-T cells in eight patients with r/r B-NHL have resulted in an 87.5% CR rate and durable responses for 1 year without grade 3 toxicity. This treatment has outperformed LV-19BBζ CAR-T cells [128]. Although genetic PD1 gene deletion reverses immunosuppression by PD-1 ligands [129], long-lasting PD-1 mRNA knockdown by stable transgenic expression of PD-1 targeted short hairpin (sh) RNA decreases surface PD-1 on CD19 CAR-T cells and impairs long-term anti-tumor function, particularly at lower ratios of effector cells to target cells. Similarly, an anti-PD1 blocking mAb accelerates CAR-T cell early differentiation and maturation, attenuates proliferation, and diminishes survival [130, 131]. Different from targeting CAR to the PD-1 locus, CC chemokine receptor 5 (CCR5) locus, adeno-associated virus site 1 (AAVS1) locus, and ß2M locus, targeting CAR to the TCRα chain constant region (TRAC) locus disrupted endogenous TCR, enhanced T cell potency after engineering. Specifically, integration of CAR to the TRAC locus averts 1928ζ CAR-T cells tonic signaling, delays 1928ζ CAR-T cells differentiation and exhaustion through optimal regulation of surface CAR levels after repetitive antigen stimulation. Surface 1928ζ CAR at the TRAC locus are downregulated within 12 hours (h) of antigen binding. The density of surface 1928ζ CAR in T cells gradually increased at 24 h after initial CD19 antigen binding. Recyclable 1928ζ CAR at the TRAC locus through the endocytosis pathway favored less-exhausted TRAC-1928ζ CAR-T cells after repeated antigen exposure [128, 132]. Data demonstrated that CD8+CAR-T cell function is abrogated following endogenous TCR activation. The TRAC site-specific CAR integration minimizes competitive activation of endogenous TCR signaling on CAR-T cell [133]. Binding of cognate ligands initiates ubiquitination of lysine residues of the cytoplasmic domain of CAR and lysosomal degradation of polyubiquitinated CAR [134, 135]. To ameliorate the loss of surface CAR in LV integrated CAR-T cells, a recyclable CAR that lacks the lysine residues of the intracellular domain can block the CAR ubiquitination pathway and promote CAR recycling to the T cell surface. TRAC locus integrated CAR-T cells have a dynamic uniform CAR surface density, thereby bypassing the CAR ubiquitination pathway [136].

            2.5 Manufacturing CAR-T cells with younger phenotypes

            In addition to patient status, age, tumor burden, and tumorigenic mutations, the numbers and quality of CD3+T cells in peripheral blood during leukapheresis in an individual patient may serve as clinical biomarkers for predicting the anti-tumor efficacy of autologous CAR-T cell products [35, 137].

            Collection of CD3+T cells from adult cancer patients receiving T cell-impairing drugs, who have chronic infections and rapidly progressive disease, poses a challenge to providing good leukapheresis products before CAR-T cells production. In addition, thymus involution occurs with age and is accompanied by decreasing naïve T cell egress from thymus [138]. Clinical data have shown that a CD3+ T cell count above a threshold of 553/μL is associated with rapidly proliferating expansion kinetics in the peripheral blood 7 days after tisagenlecleucel infusion, and favorable progression free survival (PFS) and OS in patients with r/r DLBCL [139, 140]. Moreover, an increased frequency of naïve or stem-cell memory phenotype (CD3+Tscm) of CD8 T cells apheresis is independently associated with a sustained response to tisagenlecleucel [141, 142]. In contrast, senescence CD8+CD57hiCD39hiCD28low 19CAR-T cells or exhausted CD8+PD-1hiLAG3hi 19CAR-T cells are closely associated with decreased in vivo proliferative capacity and cytotoxicity in non-responders with r/r DLBCL [143]. For the treatment of adult patients with r/r LBCL, the dosage selection of axicabtagene ciloleucel, tisagenlecleucel, and Liso-cel should consider the available numbers of transduced 19CAR in CD3+ T cells of autologous final CAR-T cells product, patients’ body weight (kg), tumor burden, CAR constructs, the choice of pre-conditioning lymphodepletion regimen [144146]. Strategies are needed to increase the efficacy and achieve the necessary amounts of CAR-T cells by supplementation with targeted drugs during the CAR-T cell production process. The production of autologous CAR-T cells involves activation of CD3+ T cells, CAR delivery by viral or non-viral methods, and an expansion process to achieve the required numbers. Preparing cellular starting material before CAR-T cell manufacture, enrichment of less-differentiated Tscm cells in leukapheresis peripheral blood mononuclear cells is ideal for overcoming T cells functional exhaustion of cancer patients. However, T cells derived from most adult patients with r/r DLBCL have no active metabolic status or terminal differentiation, and may be exhausted and express detectable immune checkpoint molecules. Moreover, in vitro activation of CD3+ T cells and CAR transduction with lentiviral vectors lead to T cell differentiation and exhaustion. Numerous developments have been made to shorten the in vitro manufacturing time and achieve transient cessation of CAR-T cell activation through the selection of epigenetic or enzymatic inhibitors to produce less differentiated “young” CAR-T cells [147, 148]. T cell exhaustion is a process of epigenic modification characterized by the expression of several key transcription factors, such as TOX and Nuclear Receptor Subfamily 4 Group A Member 3 (NRA4). NRA4 suppresses T cell factor 1 (TCF1). NFAT of activated T cells also activates the TOX/NRA4 axis [149153]. In addition, Enhancer of Zeste 2 (EZH2), the catalytic subunit of the repressive Polycomb complex 2, silences memory-associated genes through trimethylation of histone 3 lysine 27 [154]. In addition, DNA methyltransferase 1 (DNMT1), DNA methyltransferase 3B (DNMT3B), and DNA methyltransferase 3A (DNMT3A) induce genome-wide de novo DNA methylation in terminally exhausted T cells [155, 156]. Recently, studies have focused on enrichment of CAR-Tscm cells or active metabolic CAR-T cells by modifying CAR-T cell transduction or expansion processes with small molecule drug inhibitors targeting epigenetic or metabolic enzymes. For example, addition of 10 nM decitabine, an inhibitor of DNA methyltransferase, synchronously with tandem CD19/CD20 CAR transduction, enriches human CAR-T cells with a memory phenotype [157]. In another case, the FDA-approved Src kinase inhibitor dasatinib targeting Lck or Fyn has been found to attenuate proximal CAR-T cell signaling and to be associated with less differentiated CAR-T cells during ex vivo expansion [148]. Similarly, TWS119, an agonist of the Wnt/β-catenin pathway, arrests effector T cell differentiation and enriches CD8+ stem-cell memory T cells (Tscm) [158]. As a cytokine supplement, the γc cytokine family member IL-15 downregulates mTOR activity and increases enzymes associated with fatty acid oxidation, thereby maintaining the metabolic state of the Tscm population [159, 160]. IL-21 acts synergistically with IL-15 during lentiviral transduction and expansion, thereby enriching functional CD8+CAR-Tcm cells [161, 162]. Moreover, overexpression of the canonical AP-1 of heterodimeric c-Jun/fos prevents exhaustion of CAR-T cells [163].

            2.6 Target cell killing mechanism of CAR-T cells

            Understanding how TAA specific CAR-T cells eliminate cancer cells could help develop better T cell engineering strategies. Ligand binding-induced CAR-scFv aggregation forms a non-classical immune synapse between CAR-T cells and tumors. The disorganized pattern of Lck, the multiple interactions between lymphocyte function-associated antigen (LFA)-1 and intercellular adhesion molecule 1 (ICAM-1), and the mechanical force at the non-classical immune synapse may mediate the effector functions of CAR-T cells, such as the secretion of cytokines and cytolytic enzymes into targeted cancer cells. The kinase Lck mediates abundant CD3ζ phosphorylation, and stimulates faster and higher basal activation of CD28CD3ζ CAR-T cells. THEMIS-SHP1 counteracts the extent of CD3ζ phosphorylation and mild activation of 4-1BBCD3ζ CAR-T cells in non-classical immune synapses [87]. Granzymes initiate the caspase-dependent mitochondrial apoptosis pathway and caspase-independent permeabilization of the mitochondrial outer membrane. In contrast, disruption of the nonclassical immune synapse-dependent release of perforin and the granzyme pathway is one mode of resistance to CAR-T cell therapy. The interaction of CD58−/− r/r DLBCL with CD2+19CAR-T cells is lost, thereby weakening the formation of non-classical immune synapses with cancer cells, and helping cancer cells escape killing by 19CAR-T cells [164, 165]. Similarly, in IFNγR1−/− glioblastoma, ICAM is downregulated at the transcriptional level. ICAMlo IFNγR1-/- glioblastoma failed to form a non-classical immune synapse with LFA1+ CAR-T cells, and thus glioblastoma escaped CAR-T cell killing [166]. Moreover, the Fas ligand homotrimer of activated CD8+CAR-T cells mediates the trimerization of the Fas (death) receptor in cancer cells and leads to apoptosis. Similarly, Fc-receptor-activated CAR-T cells of the extracellular spacer domain of IgG CH2CH3 lead to AICD in vivo through the Fas ligand-Fas receptor axis. Chronic TAA exposure and repeated stimulation induce AICD of CAR-T cells [71, 167]. In contrast, cancer cells with mutagenesis of Fas associated death domain (FADD), BH3-interacting-domain death agonist (BID), caspase 8, and TNF receptor superfamily member 10B (death receptor 5) escape apoptosis in the presence of CAR-T cells [168, 169]. In particular, NOXA, a protein of the Bcl2 family, has been identified as a central regulator of resistance to CAR-T cells in r/r DLBCL. NOXA is potential biomarker for r/r DLBCL non-response [170]. Moreover, TNF-associated apoptosis-inducing ligands (TRAIL and CD253) selectively trigger apoptosis of tumor cells expressing TRAIL death receptors (DR). For example, TRAIL-R1 (DR4) specific CAR-T cells have shown cross cancer antitumor efficacy in preclinical studies. The combination of TRAIL-R2 (DR5) CAR-T cells targeting myeloid-derived suppressor cell (MDSC) and MUC1-specific CAR-T cells against breast cancer have shown superior anti-tumor potential against breast tumors [171, 172]. Moreover, caspase 3-mediated gasdermin E-dependent pyroptosis in B-cell cancers is associated with cytokine release syndrome [21, 173] ( Table 1 ).

            Table 1 |

            Multiple mechanisms used by CAR-T cell to eliminate targeted cancer cell.

            3. Challenges and solutions of autologous second-generation CD19 specific CAR-T cells therapy in r/r DLBCL

            Second-generation CD19-specific CAR-T cells promised a good clinical outcome in approximately 43% of treated r/r DLBCL patients within one year. However, to increase the clinical response rate for those non-responders, T-cell engineering strategies have been developed. To overcome low TAA density tumors, immunosuppressive TME and CAR-T cell exhaustion, the development of bispecific tandem or loop CAR recognition domains, human leukocyte antigen-independent synthetic TCRαβ double-chain receptors integrated into the constant region of the TCRα chain, and armored CAR-T cells were presented in the following sections ( Figure 2 ).

            Figure 2 |

            Challenges and solutions in CAR-T cell therapy for r/r DLBCL.

            The illustrated chart lists demonstrate three types of challenges in the current CAR-T cell therapy for the treatment of patients with r/r DLBCL. The proposed solutions are indicated. MDSC, myeloid derived suppressor cell; TAN, tumor associated neutrophils; TAM, tumor associated macrophages; TRAC, TCRα chain constant region; HIT, human leukocyte antigen independent TCR αβ receptor.

            3.1 Bispecific CAR-T cells

            Dual antigen specific CAR-T cells have efficacy and clinical safety for patients with CD19+ and/or CD19−/low relapse, or native B cell cancers co-expressing two TAAs [174, 175]. Bispecific CAR-T cells have been clinically applied in several strategies, including the simultaneous transduction of two separate CAR vectors into T cells or the use of a bicistronic CAR construct for expression of two separate CARs in a single vector, the parallel-production of two separate antigen-specific CAR-T cells, administered sequentially within a day (cocktail CAR-T cells), or the sequential infusion of two antigen-specific CAR-T cells within a short time interval. Notably, after undetectable CD19BBζ CAR-T cells in peripheral blood of the patient, sequential administration of humanized CD22BBζ CAR-T cells within short time intervals promotes expansion of initial shrinking CD19BBζ CAR-T cells in the r/r B-NHL patients [176]. Moreover, a cocktail of CD22-28BBζ CAR-T cells and CD19-28BBζ CAR-T cells has been found to result in an ORR of 72.2% and 50% CR after treatment of 38 patients with r/r B-NHL. The median PFS was 9.9 months, and the median OS was 18.0 months after a median follow-up of 14.4 months [177]. In addition, the combination of high-dose chemotherapy, autologous stem cell transplantation, and a cocktail of CD22-28BBζ CAR-T cells and CD19-28BBζ CAR-T cells has been found to achieve a CR of 83.3% in 42 patients with aggressive r/r DLBCL after a median follow-up of 24.3 months [178].

            In the development of a dual scFv in a single CAR design, product development of bispecific CAR-T cells could be evaluated more consistently. Bispecific tandem CARs have been developed by linking two separate scFvs in tandem. Bispecific loop CARs have been developed by linking two separate scFvs in a loop structure. The advantages of bispecific tandem or loop CAR is functional bivalency when bind to two separate TAAs of a target cell. By increasing the overall avidity of the non-classical immune synapse between the bispecific CAR-T cells and cancer targets, the “on-target” effects are increased, and the “off-target” effects are deceased. Notably, tandem CD20 (leu-16 mAb)/CD19 (FMC63 mAb) BBζ CAR-T cells have shown superior clinical safety and high efficacy in treating patients with r/r DLBCL: a 79% ORR, 71% CR, and 64% 12-month PFS have been achieved in 33 patients with r/r B-NHL in a phase I/II clinical trial. In long-term follow-up, an impressive PFS of 27.7 months was observed. Importantly, tandem CD20/CD19 BBζ CAR-T cells are associated with diminished ICAN severity [175, 179181]. Bispecific tandem CD19 (FMC63 mAb)/CD22 (humanized) BBζ CAR-T cells have been found to achieve an ORR of 87.5% and a CR of 62.5% in the treatment of 16 patients with r/r DLBCL. The 2 year OS and PFS rates were 77.3% and 40.2%, respectively, in the phase I clinical trial. Of note, 16 patients with r/r B-NHL had no ICAN and little occurrence of severe CRS [182]. Bispecific looped CD19 (FMC63 mAb), VH-CD22 (M971 mAb), and VL-CD22VH-CD19VL BBζ CAR-T cells have achieved a 62% ORR and 29% CR in 21 patients with r/r LBCL ( Table 2 ) [183].

            Table 2 |

            Comparative advantages and disadvantages of different forms of CAR-T cells targeting two tumor-associated antigens.

            Forms of CAR-T cells targeting two tumor-associated antigensAdvantagesDisadvantages
            Sequential administration or CAR-T cell cocktails by mixing of two separate antigen-specific CAR-T cells
            1. Simple product development

            2. Combinations and dosages can be flexibly adjusted according to clinical need

            1. High cost

            2. Highly variable CAR-T cell cocktails

            Generation of dual CAR-T cells through simultaneous transduction of a T cell with two different CAR vectors, thus resulting in two scFvs in a single T cell
            1. Easy product development

            1. Inconsistency of dual CAR-T cell products

            2. Challenges in pharmaceutical product control

            Bispecific tandem CAR-T cells with two distinct scFvs connected in tandem in a single CAR construct
            1. Simple product development

            2. Dual scFv design increases the functional avidity of the tandem CAR-T cells and the efficacy

            3. Safety

            1. Technical challenges in development of optimized tandem CARs

            Bicistronic CAR vectors encoding two different CARs on the same T cell
            1. Easy evolution of bicistronic CAR on the basis of the combination of two separate single-chain CAR sequences

            1. Possible homogeneous recombination between two separate single-chain CAR sequences

            2. Potential CAR-T cell tonic signaling and exhaustion because of the two copies of intracellular CD3-zeta chains

            3.2 Native TCRαβ receptor and endogenous CD3 signaling machinery

            Thymus-derived mature TCRαβ are highly sensitively to human leukocyte antigen (HLA)-loaded exogenous intracellular peptides (pep/HLA) and are approximately 100-fold more sensitive than 1928ζ CAR-T cells. Experimental data have indicated that approximately 1–50 ligands of the non-self-peptide/HLA complex per target cell triggers CD8+CTL activation signals. In addition, one to four exogenous Pep/HLA complexes per target cell triggers CD4+T cell activation. However, the presence of at least 3000 CD19 molecules per target cell triggers 1928ζ CAR intracellular signal transduction in T cells. The CD19 density of a normal B cell is ~20,000 molecules per cell. Low CD19 density (clone SJ25C1) is more common in CD10-positive DLBCL than CD10-negative nodal DLBCL samples [184]. A CD19 site-density of 952 molecules per r/r DLBCL biopsy has been found to result in CD19low relapse and progression 3 months after initial diagnostic CR [183]. Follicular lymphoma have CD19low antigen in 79% cases [185]. The decrease in CD19 antigen may be mediated by impaired membrane trafficking [186]. Trogocytosis of 19CAR-T cells has resulted in a decrease in CD19 in malignant B cells in a co-culture experiment [187]. A low CD22 density of less than 3000 molecules per malignant B cell impairs the functionality and persistence of 22BBζ CAR-T cells [59]. Furthermore, the CD20 site density is ~100,000 molecules per normal mature B cell [184]. CD20 expression below the level of 25,000 molecules per B-cell lymphoma is regarded as a low CD20 expression, and might not benefit from rituximab-based chemoimmunotherapy [188]. Native TCRαβ have a KD of 1–100 μmol/L toward foreign peptide-loaded HLA, whereas CAR-T cells have a KD of approximately 0.001 μmol/L toward the native protein epitope. Direct comparison of antigen sensitivity between CAR-T and native TCRαβ is difficult, owing to different antigen recognition models and intracellular signal activation by antigen binding [189, 190].

            TCRαβ is a non-covalent heterodimer non-covalently associated with three preferred subunit pairs of CD3γɛ, CD3δɛ, and CD3ζζ. The TCRαβ-CD3 octamer has a stoichiometry 1:1:1:1 of TCRαβ:CD3γɛ:CD3δɛ:CD3ζζ dimeric modules. Each CD3γ, CD3δ, and CD3ɛ chain has a single ITAM, and the CD3ζ chain has three tandem ITAMs. At the molecular level, thymus-derived somatic V-diverse-(D)-junction (J) recombination, and the addition and removal of nucleotides at the VD and DJ junctions in complementary determining region 3 (CDR3) of the TCR variable (V)β chain of a developing T cell clone are responsible for the immune surveillance of various exogenous Pep/HLA complexes. CDR3 of TCR-Vα is determined by V and J, as well as by the addition and removal of nucleotides at the VJ junctions of developing T cell clones [191]. CDR3 of both TCR-Vα and TCR-Vβ recognizes the specific Pep/HLA complex. CDR1 and CDR2 of both TCR-Vα and Vβ contact the side chains of HLA alleles [192]. In addition, CD4 or CD8 coreceptors are costimulatory receptors that enhance the interaction between TCRαβ and pep/HLA. The involvement of the CD8β coreceptor enhances the TCRαβ response against low-affinity Pep/HLA ligand during thymic positive selection, through the formation of a TCRαβ-coreceptor zipper [193]. In addition, eight conserved amino acid peptides of the membrane-proximal region of the TCR-α chain constant region (TCR-Cα-CPM) promote close approach to the CD8 coreceptor and antigen reactivity [194, 195]. Importantly, a homologous genetic mutation in the TCR-α constant chain (TRAC) impairs assembly and/or intracellular transport of the TCR-αβ complex, thus resulting in a lack of surface expression of the TCR-αβ complex and of thymic egress of TCR-αβ cells in patients [196].

            The presence of an immunological synapse composed of TCRαβ and pMHC complexes facilitates a cascade of proximal transmembrane signaling. The mature immunological synapse consists of the peripheral supramolecular activation cluster, as well as the central supramolecular activation cluster, which is enriched in TCRαβ, coreceptors, costimulatory receptors, inhibitory receptors, phosphorylated tyrosine kinase Lck, Src homology region 2 (SH2)-containing ζ-chain-associated protein kinase 70 (ZAP70), cytoplasmic protein tyrosine phosphatases (PTPs) of SH2-containing protein tyrosine phosphatase 2 (Shp2), and PKC-theta [197, 198]. Several molecular models have been proposed to explain how TCR-αβ binding of the non-self-peptide HLA triggers signal transduction of CD3 complex consisting of three dimers of CD3γɛ, CD3δɛ, CD3ζζ. According to the conformational change model, Pep/HLA binding triggers the movement of the proximal C-terminus of the CD3ζ transmembrane helix and the exposure of ITAMs of CD3ζ [199]. The Src-family kinases p56Lck and p59FYN phosphorylate ITAMs (YXXL/V) of CD3ζζ, CD3γɛ, and CD3δɛ. Phosphorylated ITAMs recruit several key enzymes and adaptors including Lck kinase, ZAP70, and phosphorylated adaptor of LAT, thus further amplifying the activation signal [199]. Phosphorylated LAT acts as a docking site that recruits multiple signaling proteins associated with the T-cell membrane (LAT signalosomes) and results in activation of several downstream pathways [200]. The recruitment of Lck to the intracellular tail of CD4 or CD8 coreceptors, and the cyclic interaction of CD4 or CD8 with the β2-domain of HLA-DR or the α3-domain of HLA-A2 amplify the T-cell response to foreign antigens by a million-fold [201]. In addition, CD3ζ K33 polyubiquitination mediated by the E3 ligase Cbi-b and Itch counteracts the phosphorylation of CD3ζ of activated T cells via the endocytic pathway [134]. CARs are designed to be independent of endogenous TCR/CD3 signaling. Activated 1928ζ CAR-T cells have fewer phosphorylated linkers for activated T cells (LAT) in comparison with canonical TCR signaling. LAT phosphorylation is not detected in activated 19BBζ CAR-T cells [96, 202]. Increasing evidence indicates that enhancement of intracellular signaling strength after antigen recognition increases the antigen sensitivity of CAR-T cells [96, 203]. Low density tumor antigen sensitivity of 19BBζ CAR-T cells is enhanced by activation of the phosphorylated LAT-signaling pathway through the insertion of the CD3ɛ signal sequence [96].

            3.3 Human leukocyte antigen-independent TRAC locus integrated synthetic TCRαβ

            To eliminate tumor cells with low-density antigens, an ultra-sensitive human leukocyte antigen independent synthetic TCRαβ receptor (HIT) has been constructed by replacing the endogenous TCR-Vβ and TCR-Vα with VH and VL of scFv, respectively. The synthetic chimeric VHCβ and VLCα heterodimer was integrated and expressed in the TRAC locus (TRAC-HIT) of recipient T cells by CRISPR/Cas9/sgRNA and rAAV donor template. Immunoglobulin VH and VL of TRAC-HIT directs surface antigens recognition of target cell, and CD3γɛ, CD3δɛ, CD3ζζ three dimers-mediated signaling in TRAC-HIT T cells. Moreover, the TRAC-HIT-receptors provide higher antigen sensitivity than that of TRAC-28ζ CAR-T cells. Experimental data show that as few as 20 surface CD19 molecules per B-cell cancer can be recognized and killed by TRAC-HIT-receptors modified T cells after phosphorylation of extracellular signal regulated kinase (pERK), CD3ζ, CD3ζ associated protein kinase (ZAP70), and degranulation. In contrast, TRAC-28ζ CAR-T cells can kill target cells carrying 200 CD19 molecules. Similarly, low abundance target of approximal 370 surface CD70 molecules per acute myeloid leukemia (AML) could be efficiently eliminated by CD70-knockout CD70 specific TRAC-HIT T cells. Moreover, constitutive expression of the CD80/4-1BB ligand in CD70-knockout CD70 specific TRAC-HIT T cells significantly improved its in vivo survival and anti-AML potency compared with CD70-knockout CD70 specific TRAC-CAR-T cells [204].

            In contrast to the construction of the TRAC-HIT receptor, the mutant synthetic T-cell receptor and antigen receptor (mutSTAT) has been constructed by grafting the endogenous TCR-Vβ and TCR-Vα with VL and VH of scFv, and replacing the human TCR-Cβ and TCR-Cα sequences with corresponding murine sequences. Furthermore, an additional disulfide bond was introduced between the chains in the murine (m) TCR-Cβ(S57C) and mTCR-Cα (S48C), and a hydrophobic sequence was introduced into the transmembrane domain of mTCR-Cα (S48C) to improve surface display and achieve proper pairing between VH-mTCR-Cα (S48C) and VL-mTCR-CαCβ (S57C) chains. In addition, the mTCR-Cα-connecting peptide of mutSTAT function in correctly pairing of TCR/CD3 complex after T cell engineering. Lentiviral vector-mediated transduction of mutSTAT is effective against solid cancers, particularly solid cancer cells with low antigen density, which outperform 28ζ CAR-T cells. In the resting state, mutSTAT has lower tonic signaling than 28ζ CAR-T cells [205]. Moreover, mutSTAT co-stimulated with OX40 significantly prolongs the in vivo persistence. CD19 specific mutSTAR-OX40 has been found to have a 100% CR rate in treating 18 patients with r/r B-ALL 4 weeks post infusion in a phase I clinical trial: 75% (12/16) patients remained leukemia-free after a median follow-up of 545 (433–665) days; 55.6% patients (10/18) had mild CRS; and two patients had grade III neurotoxicity [206]. In another preclinical study, VL and VH domain of the Fab of human anti-CD19 (ET190L1, clone) were fused with the δ chain and γ chain of TCR respectively to direct CD19 antigen specificity and antigen dependent CD3γɛ/CD3δɛ/CD3ζζ signaling, cytokine production, degranulation. Significantly, Fab antibody TCRγδ engineered T cells had less differentiated and exhausted Tscm phenotype, and a lower CRS than 19CAR-T cells in patients derived xenograft leukemia model [207]. In another construct design, a TCR fusion construct (TRuCs) contained an anti-CD19 scFv bound to one of the full-length TCR/CD3 octamer subunits, including TCR-Vα, TCR-Vβ, CD3γ, CD3δ, or CD3ɛ, via a flexible glycine serine linker. TRuCs became an integral component of the TCR-CD3 octamer and did not affect the Pep/HLA-mediated T cell response. Although TRuC-T and CAR-T differ in the quality of intracellular signaling, TRuC-T elicits a potent anti-tumor response in both hematologic and solid cancer xenograft mouse models [208].

            3.4 Targeting the immunosuppressive tumor microenvironment

            Immune checkpoints, tumor stroma, immunosuppressive cells, hypoxia, and metabolic limitations in the TME of r/r DLBCL hinder CAR-T cell therapy [209]. Infiltration of the TME with immunosuppressive CD4+T cells, exhausted CD8+T cells, myeloid cells, normal B cells, cancer-associated fibroblasts (CAFs), chronic hepatitis B viral infection, and potential cell-cell interaction between malignant B cells and tumor-infiltrating cells promote tumor cell survival and immune invasion [210, 211]. Moreover, activation of hypoxia-inducible factor 1 alpha in r/r DLBCL upregulates the enzyme hexokinase 2, thus promoting glucose metabolism and facilitating tumor growth under hypoxic stress. However, under hypoxic stress (<2% O2 concentrations), expansion, cytokine production, and granzyme B release of second-generation CAR-T cells are markedly diminished. Decreases in protein synthesis and defective CAR-T cell metabolism in TME have been reported [212214]. Hypoxia-activated CAR-T cells have been engineered to adapt and survive in the TME [215]. CAFs promote r/r DLBCL survival, angiogenesis, and cancer niche formation. CAFs upregulate the expression of fibroblast activation protein (FAP), smooth muscle α-actin (αSMA), programmed cell death ligand 1 (PD-L1), and PD-L2, thereby suppressing PD1+CAR-T cells [216]. Patients with r/r DLBCL expressing PD-L1 have shown poor overall survival [217]. Hyaluronidase and checkpoint blockers against anti-PD-L1 manipulated CAR-T cells degrade hyaluronic acid in the extracellular matrix, and facilitate deep CAR-T cell invasion in the TME [218]. In addition, resident endothelial cells and fibroblastic reticular cells upregulate PD-L1, thus transforming lymph node podoplanin+ fibroblastic reticular cells into CAF-like immunosuppressive cells. Armored CAR-T cells secreting PD-1 blocking scFv have been engineered to overcome PD-L1 mediated immune suppression [219]. Data have indicated that infiltration of tumor associated macrophages from r/r DLBCL TME is negatively associated with remission status after 19CAR-T cell therapy [220]. In addition, CD11b+HLA-DR myeloid derived suppressor cells are associated with poor PFS and OS of 19CAR-T cells [221]. Metastasis of B cell lymphoma is associated with the epithelial-to-mesenchymal transition process [222]. Hyaluronan synthase 2 (HAS2), and tumor growth factor β (TGFβ) regulate epithelial-to-mesenchymal transition [223] via phosphorylation of SMAD1/5 [224]. Overcoming the tumor suppressor TGFβ by constructing both TGFβ- and TAA-responsive CAR-T cells may expand the therapeutic window of r/r DLBCL [225, 226]. In addition, limited trafficking and infiltration of CAR-T cells into the TME are challenges, particularly for patients with extranodal lesions of r/r DLBCL in soft tissue [227, 228]. Modification of CAR-T cells expressing specific chemokines directs CAR-T cells into the TME. Local injection of 19CAR-T cells in combination with intravenous infusion and radiotherapy before 19CAR-T cell therapy are effective alternative methods [229, 230]. PETN-deficient r/r DLBCL are infiltrated with a large numbers of myeloid derived suppressor cells (MDSC), Tregs, and abundant expression of indoleamine 2,3-dioxygenase (IDO1) protein, an immunosuppressive enzyme involved in a rate-limiting step of tryptophan catabolism. PETN-deficient r/r DLBCL exhibited reduced infiltration of CD8+CTL and NK cells compared with suppressive immune cells, which establish immunosuppressive TME in r/r DLBCL after PETN-deficiency [231]. To activate the immune response, peptide vaccines, oncolytic virus, and Toll like receptor agonists have been developed to recruit T cells into the TME in r/r DLBCL. A peptide cocktail vaccine consisting of a 21 amino acid peptide from IDO (DTLLKALLEIASCLEKALQVF) and a 19 amino acid peptide from PD-L1 (FMTYWHLLNAFTVTVPKDL), together with nivolumab (anti-PD-1), has achieved an ORR of 80% and 43% CR with a median PFS of 26 months. CD4+ T cells and CD8+ T cells have been found to infiltrate into tumor sites in responder patients [232].

            4. CONCLUSION

            CD19 specific CAR-T cells do not induce clinical remission in approximately 57% of patients with relapsed and/or refractory DLBCL. In tumors with TAA heterogeneity especially low density surface expression of TAA, the immunosuppressive tumor microenvironment poses challenges for the current CAR-T cell therapies. Moreover, CAR-T cell exhaustion under an immunosuppressive tumor microenvironment TME potentially limit its long-term efficacy. With the rapid development of CAR design, and novel T cell engineering strategies, evidences have accumulated that a synthetic T cell receptor, independent of human leukocyte antigen, delay engineered T cell exhaustion, and have at least 10 fold super TAA sensitivity than CD28 co-stimulated CARs. Moreover, the development of bispecific tandem CAR-T cells or loop CAR-T cells not only maximizes clinical safety but also overcomes the heterogeneity of TAAs of cancer cell. Future autologous and/or allogeneic T cell engineering technologies are gradually being developed to target the dynamic, complicated TME and identify combinatory targets against the TME to address unmet clinical needs.

            ACKNOWLEDGEMENTS

            Thanks for Editors, reviewers, and proofreaders in the Journal of Hematology and Oncology Discovery.

            CONFLICTS OF INTEREST

            We declare no competing interests in relation to this work.

            REFERENCES

            1. Campo E, Swerdlow SH, Harris NL, Pileri S, Stein H, et al.. The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood. 2011. Vol. 117:5019–32. 2130098410.1182/blood-2011-01-293050

            2. Ersbøll J, Schultz HB, Hougaard P, Nissen NI, Hou-Jensen K. Comparison of the working formulation of non-Hodgkin’s lymphoma with the Rappaport, Kiel, and Lukes & Collins classifications. Translational value and prognostic significance based on review of 658 patients treated at a single institution. Cancer. 1985. Vol. 55:2442–58. 398674010.1002/1097-0142(19850515)55:10<2442::aid-cncr2820551024>3.0.co;2-9

            3. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, et al.. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016. Vol. 127:2375–90. 2698072710.1182/blood-2016-01-643569

            4. Casadei B, Argnani L, Morigi A, Lolli G, Broccoli A, et al.. Effectiveness of chemotherapy after anti-PD-1 blockade failure for relapsed and refractory Hodgkin lymphoma. Cancer Med. 2020. Vol. 9:7830–6. 3288137610.1002/cam4.3262

            5. Shanbhag S, Ambinder RF. Hodgkin lymphoma: a review and update on recent progress. CA Cancer J Clin. 2018. Vol. 68:116–32. 2919458110.3322/caac.21438

            6. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et al.. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012. Vol. 366:2443–54. 2265812710.1056/NEJMoa1200690

            7. Liu W, Liu J, Song Y, Wang X, Zhou M, et al.. Mortality of lymphoma and myeloma in China, 2004-2017: an observational study. J Hematol Oncol. 2019. Vol. 12:22 3083270210.1186/s13045-019-0706-9

            8. Liu W, Liu J, Song Y, Zeng X, Wang X, et al.. Burden of lymphoma in China, 2006-2016: an analysis of the Global Burden of Disease Study 2016. J Hematol Oncol. 2019. Vol. 12:115 3174450910.1186/s13045-019-0785-7

            9. Schmitz R, Wright GW, Huang DW, Johnson CA, Phelan JD, et al.. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 2018. Vol. 378:1396–407. 2964196610.1056/NEJMoa1801445

            10. Scott DW, King RL, Staiger AM, Ben-Neriah S, Jiang A, et al.. High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma morphology. Blood. 2018. Vol. 131:2060–4. 2947595910.1182/blood-2017-12-820605

            11. Crump M, Neelapu SS, Farooq U, Van Den Neste E, Kuruvilla J, et al.. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study. Blood. 2017. Vol. 130:1800–8. 2877487910.1182/blood-2017-03-769620

            12. Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, et al.. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019. Vol. 20:31–42. 3051850210.1016/S1470-2045(18)30864-7

            13. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Reagan PM, et al.. Comparison of 2-year outcomes with CAR T cells (ZUMA-1) vs salvage chemotherapy in refractory large B-cell lymphoma. Blood Adv. 2021. Vol. 5:4149–55. 3447848710.1182/bloodadvances.2020003848

            14. Locke FL, Miklos DB, Jacobson CA, Perales MA, Kersten MJ, et al.. Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N Engl J Med. 2022. Vol. 386:640–54. 3489122410.1056/NEJMoa2116133

            15. Neelapu SS, Dickinson M, Munoz J, Ulrickson ML, Thieblemont C, et al.. Axicabtagene ciloleucel as first-line therapy in high-risk large B-cell lymphoma: the phase 2 ZUMA-12 trial. Nat Med. 2022. Vol. 28:735–42. 3531484210.1038/s41591-022-01731-4

            16. Abramson JS, Gordon LI, Palomba ML, Lunning MA. Updated safety and long term clinical outcomes in TRANSCEND NHL 001, pivotal trial of lisocabtagene maraleucel (JCAR017) in R/R aggressive NHL. J Clin Oncol. 2018. Vol. 36 15_suppl:7505 10.1200/JCO.2018.36.15_suppl.7505

            17. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, et al.. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017. Vol. 377:2531–44. 2922679710.1056/NEJMoa1707447

            18. Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, et al.. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016. Vol. 8:355ra116. 2760555110.1126/scitranslmed.aaf8621

            19. Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev. 2019. Vol. 34:45–55. 3052896410.1016/j.blre.2018.11.002

            20. Hay KA, Hanafi LA, Li D, Gust J, Liles WC, et al.. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017. Vol. 130:2295–306. 2892401910.1182/blood-2017-06-793141

            21. Liu Y, Fang Y, Chen X, Wang Z, Liang X, et al.. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol. 2020. Vol. 5:eaax7969. 3195325710.1126/sciimmunol.aax7969

            22. Smith M, Dai A, Ghilardi G, Amelsberg KV, Devlin SM, et al.. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med. 2022. Vol. 28:713–23. 3528869510.1038/s41591-022-01702-9

            23. Wagner DH Jr, Stout RD, Suttles J. Role of the CD40-CD40 ligand interaction in CD4+ T cell contact-dependent activation of monocyte interleukin-1 synthesis. Eur J Immunol. 1994. Vol. 24:3148–54. 752867110.1002/eji.1830241235

            24. Faramand R, Jain M, Staedtke V, Kotani H, Bai R, et al.. Tumor microenvironment composition and severe cytokine release syndrome (CRS) influence toxicity in patients with large B-cell lymphoma treated with axicabtagene ciloleucel. Clin Cancer Res. 2020. Vol. 26:4823–31. 3266937210.1158/1078-0432.CCR-20-1434

            25. Chen Y, Li R, Shang S, Yang X, Li L, et al.. Therapeutic potential of TNFα and IL1β blockade for CRS/ICANS in CAR-T therapy via ameliorating endothelial activation. Front Immunol. 2021. Vol. 12:623610. 3409351910.3389/fimmu.2021.623610

            26. Wei J, Liu Y, Wang C, Zhang Y, Tong C, et al.. The model of cytokine release syndrome in CAR T-cell treatment for B-cell non-Hodgkin lymphoma. Signal Transduct Target Ther. 2020. Vol. 5:134 3272803510.1038/s41392-020-00256-x

            27. Le RQ, Li L, Yuan W, Shord SS, Nie L, et al.. FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist. 2018. Vol. 23:943–7. 2962269710.1634/theoncologist.2018-0028

            28. Nicholson IC, Lenton KA, Little DJ, Decorso T, Lee FT, et al.. Construction and characterisation of a functional CD19 specific single chain Fv fragment for immunotherapy of B lineage leukaemia and lymphoma. Mol Immunol. 1997. Vol. 34:1157–65. 956676310.1016/s0161-5890(97)00144-2

            29. Stein AM, Grupp SA, Levine JE, Laetsch TW, Pulsipher MA, et al.. Tisagenlecleucel model-based cellular kinetic analysis of chimeric antigen receptor-T cells. CPT Pharmacometrics Syst Pharmacol. 2019. Vol. 8:285–95. 3084808410.1002/psp4.12388

            30. Zi FM, Ye LL, Zheng JF, Cheng J, Wang QM. Using JAK inhibitor to treat cytokine release syndrome developed after chimeric antigen receptor T cell therapy for patients with refractory acute lymphoblastic leukemia: a case report. Medicine (Baltimore). 2021. Vol. 100:e25786. 3410661310.1097/MD.0000000000025786

            31. Bachy E, Le Gouill S, Di Blasi R, Sesques P, Manson G, et al.. A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma. Nat Med. 2022. Vol. 28:2145–54. 3613815210.1038/s41591-022-01969-y

            32. Jacobson CA, Locke F, Ghobadi A, Miklos DB, Lekakis LJ, et al.. Long-term (5 year) overall survival in Zuma-1, the pivotal study of axicabtagene ciloleucel (axi-cel) in patients with refractory large B-cell lymphoma (LBCL)2022 Tandem Meetings | Transplantation & Cellular Therapy Meetings of ASTCT and CIBMTR; 2022

            33. Chow VA, Gopal AK, Maloney DG, Turtle CJ, Smith SD, et al.. Outcomes of patients with large B-cell lymphomas and progressive disease following CD19-specific CAR T-cell therapy. Am J Hematol. 2019. Vol. 94:E209–E213. 3105676210.1002/ajh.25505

            34. Nastoupil LJ, Jain MD, Feng L, Spiegel JY, Ghobadi A, et al.. Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US Lymphoma CAR T Consortium. J Clin Oncol. 2020. Vol. 38:3119–28. 3240163410.1200/JCO.19.02104

            35. Vercellino L, Di Blasi R, Kanoun S, Tessoulin B, Rossi C, et al.. Predictive factors of early progression after CAR T-cell therapy in relapsed/refractory diffuse large B-cell lymphoma. Blood Adv. 2020. Vol. 4:5607–15. 3318089910.1182/bloodadvances.2020003001

            36. Plaks V, Rossi JM, Chou J, Wang L, Poddar S, et al.. CD19 target evasion as a mechanism of relapse in large B-cell lymphoma treated with axicabtagene ciloleucel. Blood. 2021. Vol. 138:1081–85. 3404152610.1182/blood.2021010930

            37. Rabilloud T, Potier D, Pankaew S, Nozais M, Loosveld M, et al.. Single-cell profiling identifies pre-existing CD19-negative subclones in a B-ALL patient with CD19-negative relapse after CAR-T therapy. Nat Commun. 2021. Vol. 12:865 3355854610.1038/s41467-021-21168-6

            38. Zhang Z, Chen X, Tian Y, Li F, Zhao X, et al.. Point mutation in CD19 facilitates immune escape of B cell lymphoma from CAR-T cell therapy. J Immunother Cancer. 2020. Vol. 8:e001150. 3302398110.1136/jitc-2020-001150

            39. Zhou Z, Gong Q, Lin Z, Wang Y, Li M, et al.. Emerging roles of SRSF3 as a therapeutic target for cancer. Front Oncol. 2020. Vol. 10:577636. 3307261010.3389/fonc.2020.577636

            40. Fischer J, Paret C, El Malki K, Alt F, Wingerter A, et al.. CD19 isoforms enabling resistance to CART-19 immunotherapy are expressed in B-ALL patients at initial diagnosis. J Immunother. 2017. Vol. 40:187–95. 2844126410.1097/CJI.0000000000000169

            41. Perna F, Sadelain M. Myeloid leukemia switch as immune escape from CD19 chimeric antigen receptor (CAR) therapy. Transl Cancer Res. 2016. Vol. 5 Suppl 2:S221–5p. 2882485110.21037/tcr.2016.08.15

            42. Thompson EC, Cobb BS, Sabbattini P, Meixlsperger S, Parelho V, et al.. Ikaros DNA-binding proteins as integral components of B cell developmental-stage-specific regulatory circuits. Immunity. 2007. Vol. 26:335–44. 1736330110.1016/j.immuni.2007.02.010

            43. Treiber N, Treiber T, Zocher G, Grosschedl R. Structure of an Ebf1: DNA complex reveals unusual DNA recognition and structural homology with Rel proteins. Genes Dev. 2010. Vol. 24:2270–5. 2087673210.1101/gad.1976610

            44. Majzner RG, Rietberg SP, Labanieh L, Sotillo E, Weber EW, et al.. Low CD19 antigen density diminishes efficacy of CD19 CAR T cells and can be overcome by rational redesign of CAR signaling domains. Blood. 2018. Vol. 132:963 10.1182/blood-2018-99-115558

            45. Walker AJ, Majzner RG, Zhang L, Wanhainen K, Long AH, et al.. Tumor antigen and receptor densities regulate efficacy of a chimeric antigen receptor targeting anaplastic lymphoma kinase. Mol Ther. 2017. Vol. 25:2189–201. 2867634210.1016/j.ymthe.2017.06.008

            46. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, et al.. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009. Vol. 114:1537–44. 1942372810.1182/blood-2008-12-195792

            47. Cherkassky L, Morello A, Villena-Vargas J, Feng Y, Dimitrov DS, et al.. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016. Vol. 126:3130–44. 2745429710.1172/JCI83092

            48. Jain MD, Ziccheddu B, Coughlin CA, Faramand R, Griswold AJ, et al.. Whole-genome sequencing reveals complex genomic features underlying anti-CD19 CAR T-cell treatment failures in lymphoma. Blood. 2022. Vol. 140:491–503. 3547684810.1182/blood.2021015008

            49. Batista FD, Neuberger MS. Affinity dependence of the B cell response to antigen: a threshold, a ceiling, and the importance of off-rate. Immunity. 1998. Vol. 8:751–9. 965548910.1016/s1074-7613(00)80580-4

            50. Greenman R, Pizem Y, Haus-Cohen M, Goor A, Horev G, et al.. Shaping functional avidity of CAR T cells: affinity, avidity, and antigen density that regulate response. Mol Cancer Ther. 2021. Vol. 20:872–84. 3364910610.1158/1535-7163.MCT-19-1109

            51. Liu X, Jiang S, Fang C, Yang S, Olalere D, et al.. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 2015. Vol. 75:3596–607. 2633016610.1158/0008-5472.CAN-15-0159

            52. Majzner RG, Rietberg SP, Sotillo E, Dong R, Vachharajani VT, et al.. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 2020. Vol. 10:702–23. 3219322410.1158/2159-8290.CD-19-0945

            53. June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018. Vol. 359:1361–5. 2956770710.1126/science.aar6711

            54. Pan J, Tan Y, Wang G, Deng B, Ling Z, et al.. Donor-derived CD7 chimeric antigen receptor T cells for T-cell acute lymphoblastic leukemia: first-in-human, phase I trial. J Clin Oncol. 2021. Vol. 39:3340–51. 3432439210.1200/JCO.21.00389

            55. Zhang M, Yang L, Fu X, Zhang L, Meng H, et al.. First-in-human clinical trial of the autologous CD7-CART for relapsed/refractory ACUTE lymphoblastic leukemia/lymphoma. J Clin Oncol. 2020. Vol. 38 15 suppl:3026 10.1200/JCO.2020.38.15_suppl.3026

            56. Klesmith JR, Wu L, Lobb RR, Rennert PD, Hackel BJ. Fine epitope mapping of the CD19 extracellular domain promotes design. Biochemistry. 2019. Vol. 58:4869–81. 3170290910.1021/acs.biochem.9b00808

            57. Landoni E, Fucá G, Wang J, Chirasani VR, Yao Z, et al.. Modifications to the framework regions eliminate chimeric antigen receptor tonic signaling. Cancer Immunol Res. 2021. Vol. 9:441–53. 3354722610.1158/2326-6066.CIR-20-0451

            58. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, et al.. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015. Vol. 21:581–90. 2593906310.1038/nm.3838

            59. Ramakrishna S, Highfill SL, Walsh Z, Nguyen SM, Lei H, et al.. Modulation of target antigen density improves CAR T-cell functionality and persistence. Clin Cancer Res. 2019. Vol. 25:5329–41. 3111007510.1158/1078-0432.CCR-18-3784

            60. Singh N, Frey NV, Engels B, Barrett DM, Shestova O, et al.. Antigen-independent activation enhances the efficacy of 4-1BB-costimulated CD22 CAR T cells. Nat Med. 2021. Vol. 27:842–50. 3388889910.1038/s41591-021-01326-5

            61. Mao R, Kong W, He Y. The affinity of antigen-binding domain on the antitumor efficacy of CAR T cells: moderate is better. Front Immunol. 2022. Vol. 13:1032403. 3632534510.3389/fimmu.2022.1032403

            62. Ghorashian S, Kramer AM, Onuoha S, Wright G, Bartram J, et al.. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat Med. 2019. Vol. 25:1408–14. 3147790610.1038/s41591-019-0549-5

            63. Olson ML, Mause ERV, Radhakrishnan SV, Brody JD, Rapoport AP, et al.. Low-affinity CAR T cells exhibit reduced trogocytosis, preventing rapid antigen loss, and increasing CAR T cell expansion. Leukemia. 2022. Vol. 36:1943–6. 3549019710.1038/s41375-022-01585-2

            64. Castellarin M, Sands C, Da T, Scholler J, Graham K, et al.. A rational mouse model to detect on-target, off-tumor CAR T cell toxicity. JCI Insight. 2020. Vol. 5:e136012. 3254410110.1172/jci.insight.136012

            65. Hennecke S, Cosson P. Role of transmembrane domains in assembly and intracellular transport of the CD8 molecule. J Biol Chem. 1993. Vol. 268:26607–12. 8253791

            66. Leick MB, Silva H, Scarfò I, Larson R, Choi BD, et al.. Non-cleavable hinge enhances avidity and expansion of CAR-T cells for acute myeloid leukemia. Cancer Cell. 2022. Vol. 40:494–508.e5. 3545260310.1016/j.ccell.2022.04.001

            67. Hirobe S, Imaeda K, Tachibana M, Okada N. The effects of chimeric antigen receptor (CAR) hinge domain post-translational modifications on CAR-T cell activity. Int J Mol Sci. 2022. Vol. 23:4056 3540941910.3390/ijms23074056

            68. Muller YD, Nguyen DP, Ferreira LMR, Ho P, Raffin C, et al.. The CD28-transmembrane domain mediates chimeric antigen receptor heterodimerization with CD28. Front Immunol. 2021. Vol. 12:639818. 3383375910.3389/fimmu.2021.639818

            69. Alabanza L, Pegues M, Geldres C, Shi V, Wiltzius JJW, et al.. Function of novel anti-CD19 chimeric antigen receptors with human variable regions is affected by hinge and transmembrane domains. Mol Ther. 2017. Vol. 25:2452–65. 2880756810.1016/j.ymthe.2017.07.013

            70. Guest RD, Hawkins RE, Kirillova N, Cheadle EJ, Arnold J, et al.. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother. 2005. Vol. 28:203–11. 1583837610.1097/01.cji.0000161397.96582.59

            71. Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, et al.. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res. 2015. Vol. 3:125–35. 2521299110.1158/2326-6066.CIR-14-0127

            72. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011. Vol. 365:725–33. 2183094010.1056/NEJMoa1103849

            73. Srivastava S, Riddell SR. Engineering CAR-T cells: design concepts. Trends Immunol. 2015. Vol. 36:494–502. 2616925410.1016/j.it.2015.06.004

            74. Xiao Q, Zhang X, Tu L, Cao J, Hinrichs CS, et al.. Size-dependent activation of CAR-T cells. Sci Immunol. 2022. Vol. 7:eabl3995. 3593065310.1126/sciimmunol.abl3995

            75. Ying Z, Huang XF, Xiang X, Liu Y, Kang X, et al.. A safe and potent anti-CD19 CAR T cell therapy. Nat Med. 2019. Vol. 25:947–53. 3101120710.1038/s41591-019-0421-7

            76. Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, et al.. Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature. 2022. Vol. 602:503–9. 3511073510.1038/s41586-021-04390-6

            77. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, et al.. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011. Vol. 3:95ra73. 2183223810.1126/scitranslmed.3002842

            78. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989. Vol. 86:10024–8. 251356910.1073/pnas.86.24.10024

            79. Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, et al.. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004. Vol. 18:676–84. 1496103510.1038/sj.leu.2403302

            80. Jensen MC, Popplewell L, Cooper LJ, DiGiusto D, Kalos M, et al.. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant. 2010. Vol. 16:1245–56. 2030408610.1016/j.bbmt.2010.03.014

            81. Thistlethwaite FC, Gilham DE, Guest RD, Rothwell DG, Pillai M, et al.. The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol Immunother. 2017. Vol. 66:1425–36. 2866031910.1007/s00262-017-2034-7

            82. Philipson BI, O’Connor RS, May MJ, June CH, Albelda SM, et al.. 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling. Sci Signal. 2020. Vol. 13:eaay8248. 3223496010.1126/scisignal.aay8248

            83. Buck MD, O’Sullivan D, Klein Geltink RI, Curtis JD, Chang CH, et al.. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell. 2016. Vol. 166:63–76. 2729318510.1016/j.cell.2016.05.035

            84. Kawalekar OU, O’Connor RS, Fraietta JA, Guo L, McGettigan SE, et al.. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 2016. Vol. 44:380–90. 2688586010.1016/j.immuni.2016.01.021

            85. Li G, Boucher JC, Kotani H, Park K, Zhang Y, et al.. 4-1BB enhancement of CAR T function requires NF-κB and TRAFs. JCI Insight. 2018. Vol. 3:e121322. 3023228110.1172/jci.insight.121322

            86. Gomes-Silva D, Mukherjee M, Srinivasan M, Krenciute G, Dakhova O, et al.. Tonic 4-1BB costimulation in chimeric antigen receptors impedes T cell survival and is vector-dependent. Cell Rep. 2017. Vol. 21:17–26. 2897847110.1016/j.celrep.2017.09.015

            87. Sun C, Shou P, Du H, Hirabayashi K, Chen Y, et al.. THEMIS-SHP1 recruitment by 4-1BB tunes LCK-mediated priming of chimeric antigen receptor-redirected T cells. Cancer Cell. 2020. Vol. 37:216–225.e6. 3200444110.1016/j.ccell.2019.12.014

            88. Zhao Z, Condomines M, van der Stegen SJC, Perna F, Kloss CC, et al.. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015. Vol. 28:415–28. 2646109010.1016/j.ccell.2015.09.004

            89. Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol. 1998. Vol. 161:2791–7. 9743337

            90. Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, et al.. The CD28 signaling pathway regulates glucose metabolism. Immunity. 2002. Vol. 16:769–77. 1212165910.1016/s1074-7613(02)00323-0

            91. Pollizzi KN, Sun IH, Patel CH, Lo YC, Oh MH, et al.. Asymmetric inheritance of mTORC1 kinase activity during division dictates CD8+ T cell differentiation. Nat Immunol. 2016. Vol. 17:704–11. 2706437410.1038/ni.3438

            92. Wang H, Song X, Shen L, Wang X, Xu C. Exploiting T cell signaling to optimize engineered T cell therapies. Trends Cancer. 2022. Vol. 8:123–34. 3481015610.1016/j.trecan.2021.10.007

            93. Salter AI, Ivey RG, Kennedy JJ, Voillet V, Rajan A, et al.. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci Signal. 2018. Vol. 11:eaat6753. 3013137010.1126/scisignal.aat6753

            94. Lamure S, Van Laethem F, De Verbizier D, Lozano C, Gehlkopf E, et al.. Clinical and product features associated with outcome of dlbcl patients to cd19-targeted car t-cell therapy. Cancers (Basel). 2021. Vol. 13:4279 3450308810.3390/cancers13174279

            95. Boomer JS, Green JM. An enigmatic tail of CD28 signaling. Cold Spring Harb Perspect Biol. 2010. Vol. 2:a002436. 2053470910.1101/cshperspect.a002436

            96. Salter AI, Rajan A, Kennedy JJ, Ivey RG, Shelby SA, et al.. Comparative analysis of TCR and CAR signaling informs CAR designs with superior antigen sensitivity and in vivo function. Sci Signal. 2021. Vol. 14:eabe2606. 3442938210.1126/scisignal.abe2606

            97. Faude S, Wei J, Muralidharan K, Xu X, Wertheim G, et al.. Absolute lymphocyte count proliferation kinetics after CAR T-cell infusion impact response and relapse. Blood Adv. 2021. Vol. 5:2128–36. 3388146510.1182/bloodadvances.2020004038

            98. Feucht J, Sun J, Eyquem J, Ho YJ, Zhao Z, et al.. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat Med. 2019. Vol. 25:82–8. 3055942110.1038/s41591-018-0290-5

            99. Wu W, Zhou Q, Masubuchi T, Shi X, Li H, et al.. Multiple signaling roles of CD3ɛ and its application in CAR-T cell therapy. Cell. 2020. Vol. 182:855–71.e23. 3273080810.1016/j.cell.2020.07.018

            100. Guedan S, Madar A, Casado-Medrano V, Shaw C, Wing A, et al.. Single residue in CD28-costimulated CAR-T cells limits long-term persistence and antitumor durability. J Clin Invest. 2020. Vol. 130:3087–97. 3206926810.1172/JCI133215

            101. Weinkove R, George P, Dasyam N, McLellan AD. Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin Transl Immunology. 2019. Vol. 8:e1049. 3111070210.1002/cti2.1049

            102. Goodman DB, Azimi CS, Kearns K, Talbot A, Garakani K, et al.. Pooled screening of CAR T cells identifies diverse immune signaling domains for next-generation immunotherapies. Sci Transl Med. 2022. Vol. 14:eabm1463. 3635098410.1126/scitranslmed.abm1463

            103. Ramos CA, Rouce R, Robertson CS, Reyna A, Narala N, et al.. In vivo fate and activity of second-versus third-generation CD19-specific CAR-T cells in B cell non-Hodgkin’s lymphomas. Mol Ther. 2018. Vol. 26:2727–37. 3030981910.1016/j.ymthe.2018.09.009

            104. Zhong XS, Matsushita M, Plotkin J, Riviere I, Sadelain M. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther. 2010. Vol. 18:413–20. 1977374510.1038/mt.2009.210

            105. Guedan S, Posey AD Jr, Shaw C, Wing A, Da T, et al.. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight. 2018. Vol. 3:e96976. 2932136910.1172/jci.insight.96976

            106. Muliaditan T, Halim L, Whilding LM, Draper B, Achkova DY, et al.. Synergistic T cell signaling by 41BB and CD28 is optimally achieved by membrane proximal positioning within parallel chimeric antigen receptors. Cell Rep Med. 2021. Vol. 2:100457. 3502860410.1016/j.xcrm.2021.100457

            107. Larson RC, Maus MV. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat Rev Cancer. 2021. Vol. 21:145–61. 3348371510.1038/s41568-020-00323-z

            108. Zhang H, Li F, Cao J, Wang X, Cheng H, et al.. A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity. Sci Transl Med. 2021. Vol. 13:eaba7308. 3350465110.1126/scitranslmed.aba7308

            109. Wang E, Wang LC, Tsai CY, Bhoj V, Gershenson Z, et al.. Generation of potent T-cell immunotherapy for cancer using DAP12-based, multichain, chimeric immunoreceptors. Cancer Immunol Res. 2015. Vol. 3:815–26. 2594135110.1158/2326-6066.CIR-15-0054

            110. Sun M, Xu P, Wang E, Zhou M, Xu T, et al.. Novel two-chain structure utilizing KIRS2/DAP12 domain improves the safety and efficacy of CAR-T cells in adults with r/r B-ALL. Mol Ther Oncolytics. 2021. Vol. 23:96–106. 3470387910.1016/j.omto.2021.08.014

            111. Zennou V, Petit C, Guetard D, Nerhbass U, Montagnier L, et al.. HIV-1 genome nuclear import is mediated by a central DNA flap. Cell. 2000. Vol. 101:173–85. 1078683310.1016/S0092-8674(00)80828-4

            112. Michieletto D, Lusic M, Marenduzzo D, Orlandini E. Physical principles of retroviral integration in the human genome. Nat Commun. 2019. Vol. 10:575 3071850810.1038/s41467-019-08333-8

            113. Barry SC, Harder B, Brzezinski M, Flint LY, Seppen J, et al.. Lentivirus vectors encoding both central polypurine tract and posttranscriptional regulatory element provide enhanced transduction and transgene expression. Hum Gene Ther. 2001. Vol. 12:1103–8. 1139923110.1089/104303401750214311

            114. Hasegawa Y, Nakagawa SJ. Revisiting the function of nuclear scaffold/matrix binding proteins in X chromosome inactivation. RNA Biol. 2011. Vol. 8:735–9. 2188141210.4161/rna.8.5.16367

            115. Ramezani A, Hawley RG. Strategies to insulate lentiviral vector-expressed transgenesLentivirus Gene Engineering Protocols. Springer. 2010. p. 77–100

            116. Schambach A, Bohne J, Baum C, Hermann FG, Egerer L, et al.. Woodchuck hepatitis virus post-transcriptional regulatory element deleted from X protein and promoter sequences enhances retroviral vector titer and expression. Gene Ther. 2006. Vol. 13:641–5. 1635511410.1038/sj.gt.3302698

            117. Paugh BS, Baranyi L, Roy A, He HJ, Harris L, et al.. Reference standards for accurate validation and optimization of assays that determine integrated lentiviral vector copy number in transduced cells. Sci Rep. 2021. Vol. 11:389 3343198910.1038/s41598-020-79698-w

            118. Atianand MK, Fitzgerald KA. Molecular basis of DNA recognition in the immune system. J Immunol. 2013. Vol. 190:1911–8. 2341752710.4049/jimmunol.1203162

            119. Ellis J. Silencing and variegation of gammaretrovirus and lentivirus vectors. Hum Gene Ther. 2005. Vol. 16:1241–6. 1625955710.1089/hum.2005.16.1241

            120. Lamers CH, Willemsen R, van Elzakker P, van Steenbergen-Langeveld S, Broertjes M, et al.. Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. Blood. 2011. Vol. 117:72–82. 2088992510.1182/blood-2010-07-294520

            121. Tao J, Zhou X, Jiang Z. cGAS-cGAMP-STING: the three musketeers of cytosolic DNA sensing and signaling. IUBMB Life. 2016. Vol. 68:858–70. 2770689410.1002/iub.1566

            122. Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, et al.. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018. Vol. 558:307–12. 2984914110.1038/s41586-018-0178-z

            123. Nobles CL, Sherrill-Mix S, Everett JK, Reddy S, Fraietta JA, et al.. CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration. J Clin Invest. 2020. Vol. 130:673–85. 3184590510.1172/JCI130144

            124. Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 2014. Vol. 157:1262–78. 2490614610.1016/j.cell.2014.05.010

            125. Salsman J, Dellaire G. Precision genome editing in the CRISPR era. Biochem Cell Biol. 2017. Vol. 95:187–201. 2817777110.1139/bcb-2016-0137

            126. Kovacˇ A, Miskey C, Menzel M, Grueso E, Gogol-Döring A, et al.. RNA-guided retargeting of sleeping beauty transposition in human cells. Elife. 2020. Vol. 9:e53868. 3214240810.7554/eLife.53868

            127. Querques I, Mades A, Zuliani C, Miskey C, Alb M, et al.. A highly soluble sleeping beauty transposase improves control of gene insertion. Nat Biotechnol. 2019. Vol. 37:1502–12. 3168595910.1038/s41587-019-0291-z

            128. Zhang J, Hu Y, Yang J, Li W, Zhang M, et al.. Non-viral, specifically targeted CAR-T cells achieve high safety and efficacy in B-NHL. Nature. 2022. Vol. 609:369–74. 3604529610.1038/s41586-022-05140-y

            129. Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, et al.. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep. 2017. Vol. 7:737 2838966110.1038/s41598-017-00462-8

            130. Kalinin R, Ukrainskaya VM, Chumakov SP, Moysenovich AM, Tereshchuk VM, et al.. Engineered removal of PD-1 from the surface of CD19 CAR-T cells results in increased activation and diminished survival. Front Mol Biosci. 2021. Vol. 8:745286. 3472263310.3389/fmolb.2021.745286

            131. Wei J, Luo C, Wang Y, Guo Y, Dai H, et al.. PD-1 silencing impairs the anti-tumor function of chimeric antigen receptor modified T cells by inhibiting proliferation activity. J Immunother Cancer. 2019. Vol. 7:209 3139109610.1186/s40425-019-0685-y

            132. Eyquem J, Mansilla-Soto J, Giavridis T, van der Stegen SJ, Hamieh M, et al.. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017. Vol. 543:113–7. 2822575410.1038/nature21405

            133. Yang Y, Kohler ME, Chien CD, Sauter CT, Jacoby E, et al.. TCR engagement negatively affects CD8 but not CD4 CAR T cell expansion and leukemic clearance. Sci Transl Med. 2017. Vol. 9:eaag1209. 2916739210.1126/scitranslmed.aag1209

            134. Huang H, Jeon MS, Liao L, Yang C, Elly C, et al.. K33-linked polyubiquitination of T cell receptor-zeta regulates proteolysis-independent T cell signaling. Immunity. 2010. Vol. 33:60–70. 2063765910.1016/j.immuni.2010.07.002

            135. Yang M, Chen T, Li X, Yu Z, Tang S, et al.. K33-linked polyubiquitination of Zap70 by Nrdp1 controls CD8+ T cell activation. Nat Immunol. 2015. Vol. 16:1253–62. 2639015610.1038/ni.3258

            136. Li W, Qiu S, Chen J, Jiang S, Chen W, et al.. Chimeric antigen receptor designed to prevent ubiquitination and downregulation showed durable antitumor efficacy. Immunity. 2020. Vol. 53:456–70.e6. 3275841910.1016/j.immuni.2020.07.011

            137. Locke FL, Rossi JM, Neelapu SS, Jacobson CA, Miklos DB, et al.. Tumor burden, inflammation, and product attributes determine outcomes of axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv. 2020. Vol. 4:4898–911. 3303533310.1182/bloodadvances.2020002394

            138. Palmer DB. The effect of age on thymic function. Front Immunol. 2013. Vol. 4:316 2410948110.3389/fimmu.2013.00316

            139. Korell F, Laier S, Sauer S, Veelken K, Hennemann H, et al.. Current challenges in providing good leukapheresis products for manufacturing of CAR-T cells for patients with relapsed/refractory NHL or ALL. Cells. 2020. Vol. 9:1225 3242918910.3390/cells9051225

            140. Wada F, Jo T, Arai Y, Kitawaki T, Mizumoto C, et al.. T-cell counts in peripheral blood at leukapheresis predict responses to subsequent CAR-T cell therapy. Sci Rep. 2022. Vol. 12:18696. 3633352110.1038/s41598-022-23589-9

            141. Cuffel A, Allain V, Faivre L, Di Blasi R, Morin F, et al.. Real-world characteristics of T-cell apheresis and clinical response to tisagenlecleucel in B-cell lymphoma. Blood Adv. 2022. Vol. 6:4657–60. 3572805110.1182/bloodadvances.2022007057

            142. Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, et al.. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018. Vol. 24:563–71. 2971308510.1038/s41591-018-0010-1

            143. Beider K, Itzhaki O, Schachter J, Grushchenko-Polaq AH, Voevoda-Dimenshtein V, et al.. Molecular and functional signatures associated with CAR T cell exhaustion and impaired clinical response in patients with B cell malignancies. Cells. 2022. Vol. 11:1140 3540670310.3390/cells11071140

            144. Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, et al.. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020. Vol. 396:839–52. 3288840710.1016/S0140-6736(20)31366-0

            145. Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, et al.. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016. Vol. 30:492–500. 2636998710.1038/leu.2015.247

            146. Teoh J, Brown LF. Developing lisocabtagene maraleucel chimeric antigen receptor T-cell manufacturing for improved process, product quality and consistency across CD19+ hematologic indications. Cytotherapy. 2022. Vol. 24:962–73. 3561008910.1016/j.jcyt.2022.03.013

            147. Ghassemi S, Durgin JS, Nunez-Cruz S, Patel J, Leferovich J, et al.. Rapid manufacturing of non-activated potent CAR T cells. Nat Biomed Eng. 2022. Vol. 6:118–28. 3519068010.1038/s41551-021-00842-6

            148. Weber EW, Parker KR, Sotillo E, Lynn RC, Anbunathan H, et al.. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science. 2021. Vol. 372:eaba1786. 3379542810.1126/science.aba1786

            149. Martinez GJ, Pereira RM, Äijö T, Kim EY, Marangoni F, et al.. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity. 2015. Vol. 42:265–78. 2568027210.1016/j.immuni.2015.01.006

            150. Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, et al.. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019. Vol. 20:326–36. 3077825210.1038/s41590-019-0312-6

            151. Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, et al.. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018. Vol. 175:998–1013.e20. 3038845610.1016/j.cell.2018.10.038

            152. Seo H, Chen J, González-Avalos E, Samaniego-Castruita D, Das A, et al.. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc Natl Acad Sci U S A. 2019. Vol. 116:12410–5. 3115214010.1073/pnas.1905675116

            153. Seo W, Jerin C, Nishikawa H. Transcriptional regulatory network for the establishment of CD8+ T cell exhaustion. Exp Mol Med. 2021. Vol. 53:202–9. 10.1038/s12276-021-00568-0

            154. Gray SM, Amezquita RA, Guan T, Kleinstein SH, Kaech SM. Polycomb repressive complex 2-mediated chromatin repression guides effector CD8+ T cell terminal differentiation and loss of multipotency. Immunity. 2017. Vol. 46:596–608. 2841098910.1016/j.immuni.2017.03.012

            155. Ghoneim HE, Fan Y, Moustaki A, Abdelsamed HA, Dash P, et al.. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell. 2017. Vol. 170:142–57.e19. 2864866110.1016/j.cell.2017.06.007

            156. Schietinger A, Philip M, Krisnawan VE, Chiu EY, Delrow JJ, et al.. Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity. 2016. Vol. 45:389–401. 2752126910.1016/j.immuni.2016.07.011

            157. Wang Y, Tong C, Dai H, Wu Z, Han X, et al.. Low-dose decitabine priming endows CAR T cells with enhanced and persistent antitumour potential via epigenetic reprogramming. Nat Commun. 2021. Vol. 12:409 3346224510.1038/s41467-020-20696-x

            158. Gattinoni L, Zhong XS, Palmer DC, Ji Y, Hinrichs CS, et al.. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat Med. 2009. Vol. 15:808–13. 1952596210.1038/nm.1982

            159. Alizadeh D, Wong RA, Yang X, Wang D, Pecoraro JR, et al.. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol Res. 2019. Vol. 7:759–72. 3089053110.1158/2326-6066.CIR-18-0466

            160. Xu Y, Zhang M, Ramos CA, Durett A, Liu E, et al.. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood. 2014. Vol. 123:3750–9. 2478250910.1182/blood-2014-01-552174

            161. Du L, Nai Y, Shen M, Li T, Huang J, et al.. IL-21 Optimizes the CAR-T Cell Preparation Through Improving Lentivirus Mediated Transfection Efficiency of T Cells and Enhancing CAR-T Cell Cytotoxic Activities. Front Mol Biosci. 2021. Vol. 8:675179. 3417908310.3389/fmolb.2021.675179

            162. Zeng R, Spolski R, Finkelstein SE, Oh S, Kovanen PE, et al.. Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function. J Exp Med. 2005. Vol. 201:139–48. 1563014110.1084/jem.20041057

            163. Lynn RC, Weber EW, Sotillo E, Gennert D, Xu P, et al.. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature. 2019. Vol. 576:293–300. 10.1038/s41586-019-1805-z

            164. Romain G, Strati P, Rezvan A, Fathi M, Bandey IN, et al.. Multidimensional single-cell analysis identifies a role of CD2-CD58 interactions for clinical antitumor T cell responses. J Clin Invest. 2022. Vol. 132:e159402. 3588148610.1172/JCI159402

            165. Yan X, Chen D, Ma X, Wang Y, Guo Y, et al.. CD58 loss in tumor cells confers functional impairment of CAR T cells. Blood Adv. 2022. Vol. 6:5844–56. 3572806210.1182/bloodadvances.2022007891

            166. Larson RC, Kann MC, Bailey SR, Haradhvala NJ, Llopis PM, et al.. CAR T cell killing requires the IFNγR pathway in solid but not liquid tumours. Nature. 2022. Vol. 604:563–70. 3541868710.1038/s41586-022-04585-5

            167. Gargett T, Yu W, Dotti G, Yvon ES, Christo SN, et al.. 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. Vol. 24:1135–49. 2701999810.1038/mt.2016.63

            168. Singh N, Lee YG, Shestova O, Ravikumar P, Hayer KE, et al.. Impaired death receptor signaling in leukemia causes antigen-independent resistance by inducing CAR T-cell dysfunction. Cancer Discov. 2020. Vol. 10:552–67. 3200151610.1158/2159-8290

            169. Upadhyay R, Boiarsky JA, Pantsulaia G, Svensson-Arvelund J, Lin MJ, et al.. A Critical Role for Fas-Mediated Off-Target Tumor Killing in T-cell Immunotherapy. Cancer Discov. 2021. Vol. 11:599–613. 3333473010.1158/2159-8290.CD-20-0756

            170. Yan X, Chen D, Wang Y, Guo Y, Tong C, et al.. Identification of NOXA as a pivotal regulator of resistance to CAR T-cell therapy in B-cell malignancies. Sig Transduct Target Ther. 2022. Vol. 7:98 10.1038/s41392-022-00915-1

            171. Nai Y, Du L, Shen M, Li T, Huang J, et al.. TRAIL-R1-targeted CAR-T cells exhibit dual antitumor efficacy. Front Mol Biosci. 2021. Vol. 8:756599. 3498811410.3389/fmolb.2021.756599

            172. Nalawade SA, Shafer P, Bajgain P, McKenna MK, Ali A, et al.. Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer. J Immunother Cancer. 2021. Vol. 9:e003237. 3481535510.1136/jitc-2021-003237

            173. Davenport AJ, Jenkins MR, Ritchie DS, Prince HM, Trapani JA, et al.. CAR-T cells are serial killers. Oncoimmunology. 2015. Vol. 4:e1053684. 2658733010.1080/2162402X.2015.1053684

            174. Shah NN, Johnson BD, Schneider D, Zhu F, Szabo A, et al.. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat Med. 2020. Vol. 26:1569–75. 3302064710.1038/s41591-020-1081-3

            175. Tong C, Zhang Y, Liu Y, Ji X, Zhang W, et al.. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B-cell lymphoma. Blood. 2020. Vol. 136:1632–44. 3255624710.1182/blood.2020005278

            176. Meng Y, Deng B, Rong L, Li C, Song W, et al.. Short-Interval Sequential CAR-T Cell Infusion May Enhance Prior CAR-T Cell Expansion to Augment Anti-Lymphoma Response in B-NHL. Front Oncol. 2021. Vol. 11:640166. 3427740010.3389/fonc.2021.640166

            177. Wang N, Hu X, Cao W, Li C, Xiao Y, et al.. Efficacy and safety of CAR19/22 T-cell cocktail therapy in patients with refractory/relapsed B-cell malignancies. Blood. 2020. Vol. 135:17–27. 3169782410.1182/blood.2019000017

            178. Cao Y, Xiao Y, Wang N, Wang G, Huang L, et al.. CD19/CD22 Chimeric Antigen Receptor T Cell Cocktail Therapy Following Autologous Transplantation in Patients With Relapsed/Refractory Aggressive B Cell Lymphomas. Transplant Cell Ther. 2021. Vol. 27:910.e1–910.e11. 3442526010.1016/j.jtct.2021.08.012

            179. Davenport AJ, Cross RS, Watson KA, Liao Y, Shi W, et al.. Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity. Proc Natl Acad Sci U S A. 2018. Vol. 115:E2068–76. 2944040610.1073/pnas.1716266115

            180. Liu D, Badeti S, Dotti G, Jiang JG, Wang H, et al.. The role of immunological synapse in predicting the efficacy of chimeric antigen receptor (CAR) immunotherapy. Cell Commun Signal. 2020. Vol. 18:134 10.1186/s12964-020-00617-7

            181. Zhang Y, Wang Y, Liu Y, Tong C, Wang C, et al.. Long-term activity of tandem CD19/CD20 CAR therapy in refractory/relapsed B-cell lymphoma: a single-arm, phase 1–2 trial. Leukemia. 2022. Vol. 36:189–96. 3427248110.1038/s41375-021-01345-8

            182. Wei G, Zhang Y, Zhao H, Wang Y, Liu Y, et al.. CD19/CD22 Dual-Targeted CAR T-cell Therapy for Relapsed/Refractory Aggressive B-cell Lymphoma: A Safety and Efficacy Study. Cancer Immunol Res. 2021. Vol. 9:1061–70. 3429004810.1158/2326-6066.CIR-20-0675

            183. Spiegel JY, Patel S, Muffly L, Hossain NM, Oak J, et al.. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial. Nat Med. 2021. Vol. 27:1419–31. 3431255610.1038/s41591-021-01436-0

            184. Horna P, Nowakowski G, Endell J, Boxhammer R. Comparative assessment of surface CD19 and CD20 expression on B-cell lymphomas from clinical biopsies: implications for targeted therapies. Blood. 2019. Vol. 134:5345 10.1182/blood-2019-129600

            185. Yang W, Agrawal N, Patel J, Edinger A, Osei E, et al.. Diminished expression of CD19 in B-cell lymphomas. Cytometry B Clin Cytom. 2005. Vol. 63:28–35. 1562420410.1002/cyto.b.20030

            186. Braig F, Brandt A, Goebeler M, Tony HP, Kurze AK, et al.. Resistance to anti-CD19/CD3 BiTE in acute lymphoblastic leukemia may be mediated by disrupted CD19 membrane trafficking. Blood. 2017. Vol. 129:100–4. 2778467410.1182/blood-2016-05-718395

            187. Hamieh M, Dobrin A, Cabriolu A, van der Stegen SJC, Giavridis T, et al.. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature. 2019. Vol. 568:112–6. 3091839910.1038/s41586-019-1054-1

            188. Prevodnik VK, Lavrencˇak J, Horvat M, Novakovicˇ BJ. The predictive significance of CD20 expression in B-cell lymphomas. Diagn Pathol. 2011. Vol. 6:33 2148644810.1186/1746-1596-6-33

            189. Huang J, Brameshuber M, Zeng X, Xie J, Li QJ, et al.. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4+ T cells. Immunity. 2013. Vol. 39:846–57. 2412036210.1016/j.immuni.2013.08.036

            190. Irvine DJ, Purbhoo MA, Krogsgaard M, Davis MM. Direct observation of ligand recognition by T cells. Nature. 2002. Vol. 419:845–9. 1239736010.1038/nature01076

            191. Tonegawa S. Somatic generation of antibody diversity. Nature. 1983. Vol. 302:575–81. 630068910.1038/302575a0

            192. Kappler JW, Roehm N, Marrack P. T cell tolerance by clonal elimination in the thymus. Cell. 1987. Vol. 49:273–80. 349452210.1016/0092-8674(87)90568-x

            193. Palmer E, Naeher D. Affinity threshold for thymic selection through a T-cell receptor–co-receptor zipper. Nat Rev Immunol. 2009. Vol. 9:207–13. 1915174810.1038/nri2469

            194. Bäckström BT, Milia E, Peter A, Jaureguiberry B, Baldari CT, et al.. A motif within the T cell receptor α chain constant region connecting peptide domain controls antigen responsiveness. Immunity. 1996. Vol. 5:437–47. 8934571110.1016/s1074-7613(00)80500-2

            195. Mallaun M, Naeher D, Daniels MA, Yachi PP, Hausmann B, et al.. The T cell receptor’s α-chain connecting peptide motif promotes close approximation of the CD8 coreceptor allowing efficient signal initiation. J Immunol. 2008. Vol. 180:8211–21. 1852328710.4049/jimmunol.180.12.8211

            196. Morgan NV, Goddard S, Cardno TS, McDonald D, Rahman F, et al.. Mutation in the TCRα subunit constant gene (TRAC) leads to a human immunodeficiency disorder characterized by a lack of TCRαβ+ T cells. J Clin Invest. 2011. Vol. 121:695–702. 2120608810.1172/JCI41931

            197. Monks CR, Freiberg BA, Kupfer H, Sciaky N, Kupfer A. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature. 1998. Vol. 395:82–6. 973850210.1038/25764

            198. Yokosuka T, Kobayashi W, Takamatsu M, Sakata-Sogawa K, Zeng H, et al.. Spatiotemporal basis of CTLA-4 costimulatory molecule-mediated negative regulation of T cell activation. Immunity. 2010. Vol. 33:326–39. 2087017510.1016/j.immuni.2010.09.006

            199. Chen Y, Zhu Y, Li X, Gao W, Zhen Z, et al.. Cholesterol inhibits TCR signaling by directly restricting TCR-CD3 core tunnel motility. Mol Cell. 2022. Vol. 82:1278–87.e5. 3527181410.1016/j.molcel.2022.02.017

            200. Paz PE, Wang S, Clarke H, Lu X, Stokoe D, et al.. Mapping the Zap-70 phosphorylation sites on LAT (linker for activation of T cells) required for recruitment and activation of signalling proteins in T cells. Biochem J. 2001. Vol. 356:461–71. 1136877310.1042/0264-6021:3560461

            201. Mørch AM, Bálint Š, Santos AM, Davis SJ, Dustin ML. Coreceptors and TCR signaling–the strong and the weak of it. Front Cell Dev Biol. 2020. Vol. 8:597627. 3317870610.3389/fcell.2020.597627

            202. Dong R, Libby KA, Blaeschke F, Fuchs W, Marson A, et al.. Rewired signaling network in T cells expressing the chimeric antigen receptor (CAR). EMBO J. 2020. Vol. 39:e104730. 3264382510.15252/embj.2020104730

            203. Priceman SJ, Gerdts EA, Tilakawardane D, Kennewick KT, Murad JP, et al.. Co-stimulatory signaling determines tumor antigen sensitivity and persistence of CAR T cells targeting PSCA+ metastatic prostate cancer. Oncoimmunology. 2018. Vol. 7:e1380764

            204. Mansilla-Soto J, Eyquem J, Haubner S, Hamieh M, Feucht J, et al.. HLA-independent T cell receptors for targeting tumors with low antigen density. Nat Med. 2022. Vol. 28:345–52

            205. Liu Y, Liu G, Wang J, Zheng ZY, Jia L, et al.. Chimeric STAR receptors using TCR machinery mediate robust responses against solid tumors. Sci Transl Med. 2021. Vol. 13:eabb5191. 3376243710.1126/scitranslmed.abb5191

            206. Wang J, Zhang X, Zhou Z, Liu Y, Yu L, et al.. A novel adoptive synthetic TCR and antigen receptor (STAR) T-Cell therapy for B-Cell acute lymphoblastic leukemia. Am J Hematol. 2022. Vol. 97:992–1004. 3549151110.1002/ajh.26586

            207. Xu Y, Yang Z, Horan LH, Zhang P, Liu L, et al.. A novel antibody-TCR (AbTCR) platform combines Fab-based antigen recognition with gamma/delta-TCR signaling to facilitate T-cell cytotoxicity with low cytokine release. Cell Discov. 2018. Vol. 4:1–13. 10.1038/s41421-018-0066-6

            208. Baeuerle PA, Ding J, Patel E, Thorausch N, Horton H, et al.. Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response. Nat Commun. 2019. Vol. 10:2087 10.1038/s41467-019-10097-0

            209. Kim GB, Riley JL, Levine BL. Engineering T cells to survive and thrive in the hostile tumor microenvironment. Curr Opin Biomed Eng. 2022. Vol. 21:100360. 10.1016/j.cobme.2021.100360

            210. Kotlov N, et al.. Clinical and biological subtypes of B-cell lymphoma revealed by microenvironmental signatures. Cancer Discov. 2021. Vol. 11:1468–89. 3354186010.1158/2159-8290.CD-20-0839

            211. Ye X, Wang L, Nie M, Wang Y, Dong S, et al.. A single-cell atlas of diffuse large B cell lymphoma. Cell Rep. 2022. Vol. 39:110713. 3544316310.1016/j.celrep.2022.110713

            212. Berahovich R, Liu X, Zhou H, Tsadik E, Xu S, et al.. Hypoxia selectively impairs CAR-T cells in vitro. 2019. Vol. 11:602 3105226110.3390/cancers11050602

            213. Bhalla K, Jaber S, Nahid MN, Underwood K, Beheshti A, et al.. Role of hypoxia in Diffuse Large B-cell Lymphoma: Metabolic repression and selective translation of HK2 facilitates development of DLBCL. Sci Rep. 2018. Vol. 8:744 2933558110.1038/s41598-018-19182-8

            214. Gray M, Meehan J, Turnbull AK, Martínez-Pérez C, Kay C, et al.. The importance of the tumor microenvironment and hypoxia in delivering a precision medicine approach to veterinary oncology. Front Vet Sci. 2020. Vol. 7:598338. 3328293510.3389/fvets.2020.598338

            215. He H, Liao Q, Zhao C, Zhu C, Feng M, et al.. Conditioned CAR-T cells by hypoxia-inducible transcription amplification (HiTA) system significantly enhances systemic safety and retains antitumor efficacy. J Immunother Cancer. 2021. Vol. 9:e002755. 3461570410.1136/jitc-2021-002755

            216. Apollonio B, Jarvis P, Phillips B, Kuhnl A, Salisbury J, et al.. Diffuse large B-cell lymphoma remodels the fibroblastic reticular network that acquires aberrant immunosuppressive capabilities; implications for the regulation of anti-tumor immunity in the immuno-oncology era. Blood. 2018. Vol. 132:675 10.1182/blood-2018-99-116409

            217. Kiyasu J, Miyoshi H, Hirata A, Arakawa F, Ichikawa A, et al.. Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. Blood. 2015. Vol. 126:2193–201. 2623908810.1182/blood-2015-02-629600

            218. Zhao Y, Dong Y, Yang S, Tu Y, Wang C, et al.. Bioorthogonal Equipping CAR-T Cells with Hyaluronidase and Checkpoint Blocking Antibody for Enhanced Solid Tumor Immunotherapy. ACS Cent Sci. 2022. Vol. 8:603–14. 3564727410.1021/acscentsci.2c00163

            219. Rafiq S, Yeku OO, Jackson HJ, Purdon TJ, van Leeuwen DG, et al.. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol. 2018. Vol. 36:847–56. 3010229510.1038/nbt.4195

            220. Yan Z-X, Li L, Wang W, OuYang BS, Cheng S, et al.. Clinical efficacy and tumor microenvironment influence in a dose-escalation study of anti-CD19 chimeric antigen receptor T cells in refractory B-cell non-Hodgkin’s lymphoma. Clin Cancer Res. 2019. Vol. 25:6995–7003. 3144425010.1158/1078-0432.CCR-19-0101

            221. Jaeger U, Bishop MR, Salles G, Schuster SJ, Maziarz RL, et al.. Myc expression and tumor-infiltrating T cells are associated with response in patients (pts) with relapsed/refractory diffuse large B-cell lymphoma (r/r DLBCL) treated with tisagenlecleucel in the Juliet trial. Blood. 2020. Vol. 136:48–9. 10.1182/blood-2020-137045

            222. Zhang Y, Weinberg RA. Epithelial-to-mesenchymal transition in cancer: complexity and opportunities. Front Med. 2018. Vol. 12:361–73. 3004322110.1007/s11684-018-0656-6

            223. Moustakas A, Heldin P. TGFβ and matrix-regulated epithelial to mesenchymal transition. Biochim Biophys Acta. 2014. Vol. 1840:2621–34. 2456126610.1016/j.bbagen.2014.02.004

            224. Ramachandran A, Vizán P, Das D, Chakravarty P, Vogt J, et al.. TGF-β uses a novel mode of receptor activation to phosphorylate SMAD1/5 and induce epithelial-to-mesenchymal transition. Elife. 2018. Vol. 7:e31756. 2937682910.7554/eLife.31756

            225. Hou AJ, Chang ZL, Lorenzini MH, Zah E, Chen YY. TGF-β–responsive CAR-T cells promote anti-tumor immune function. Bioeng Transl Med. 2018. Vol. 3:75–86. 3006596410.1002/btm2.10097

            226. Liu Y, Zhou X, Wang X. Targeting the tumor microenvironment in B-cell lymphoma: challenges and opportunities. J Hematol Oncol. 2021. Vol. 14:1–17. 3440443410.1186/s13045-021-01134-x

            227. Foeng J, Comerford I, McColl SR. Harnessing the chemokine system to home CAR-T cells into solid tumors. Cell Rep Med. 2022. Vol. 3:100543. 3549288010.1016/j.xcrm.2022.100543

            228. Zhou L, Li P, Ye S, Tang X, Wang J, et al.. Different sites of extranodal involvement may affect the survival of patients with relapsed or refractory non-Hodgkin lymphoma after chimeric antigen receptor T cell therapy. Front Med. 2020. Vol. 14:786–91. 3278973210.1007/s11684-020-0751-3

            229. Figura NB, Sim AJ, Jain MD, Chavez JC, Robinson TJ. Radiation therapy prior to CAR T-cell therapy in lymphoma: impact on patient outcomes. Expert Rev Hematol. 2022. Vol. 15:1023–30. 3636995010.1080/17474086.2022.2147919

            230. Hu L, Wu F, Wang H, Zhu W, Wang J, et al.. Case report: combined intravenous infusion and local injection of CAR-T cells induced remission in a relapsed diffuse large B-cell lymphoma patient. Front Immunol. 2021. Vol. 12:665230. 10.3389/fimmu.2021.665230

            231. Cetintas VB, Batada NN. Is there a causal link between PTEN deficient tumors and immunosuppressive tumor microenvironment? J Transl Med. 2020. Vol. 18:45 3200079410.1186/s12967-020-02219-w

            232. Kjeldsen JW, Lorentzen CL, Martinenaite E, Ellebaek E, Donia M, et al.. A phase 1/2 trial of an immune-modulatory vaccine against IDO/PD-L1 in combination with nivolumab in metastatic melanoma. Nat Med. 2021. Vol. 27:2212–23. 3488757410.1038/s41591-021-01544-x

            Author and article information

            Journal
            hod
            Hematology and Oncology Discovery
            Compuscript (Ireland )
            2811-5619
            25 March 2023
            : 2
            : 1
            : 9-29
            Affiliations
            [a ]Cytology Laboratory, Beijing Gobroad Boren Hospital, Jitong East Road No.6, Fengtai district, Beijing 10070, China
            Author notes
            Article
            10.15212/HOD-2022-0009
            9493f087-7f80-4d70-b6a4-6d760464adae
            Copyright © 2023 The Authors.

            This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 International.

            History
            : 12 December 2022
            : 03 February 2023
            : 26 February 2023
            Page count
            Figures: 2, Tables: 2, References: 232, Pages: 21
            Categories
            Review

            Medicine,Hematology
            T cell engineering, chimeric antigen receptor (CAR),immune suppressive tumor microenvironment,synthetic TCR,low-density antigen

            Comments

            Comment on this article